1
|
Huang Y, Liu H, Zhao Y, Chen H, Li Q, Li X, Hua S, Cao D, Chang Y. Disrupting redox homeostasis for tumor therapy based on PDT/chemo/ferroptosis therapeutic hybrid liposomes. RSC Adv 2024; 14:20152-20162. [PMID: 38915327 PMCID: PMC11195642 DOI: 10.1039/d4ra03361b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/09/2024] [Indexed: 06/26/2024] Open
Abstract
Synergistic photodynamic therapy (PDT) with other therapeutic modalities can enhance the therapeutic efficacy of tumor treatment and reduce the adverse effects associated with drug leakage and off-target accumulation. However, shaping combined strategies for synergistic therapy remains challenging. Herein, we developed versatile hybrid liposomes self-assembled from Ce6-lipid conjugates and loaded with the chemo drug doxorubicin (DOX) and ferroptosis inducer Fe3O4 nanoparticles for synergistic PDT/chemo/ferroptosis therapy. Abundant ROS are generated by PDT upon 650 nm light irradiation, Fe3O4-mediated Fenton reaction, and DOX-induced apoptosis. Furthermore, amplifying oxidative stress in cancer cells to disrupt cellular redox homeostasis could accelerate tumor cell death through oxidative damage to lipids, proteins, and DNA. Overall, this work highlights liposome-based therapeutic nanoformulations, thus offering a breakthrough redox homeostasis-based synergistic PDT/chemo/ferroptosis therapy for lung cancer.
Collapse
Affiliation(s)
- Yuanping Huang
- Department of Respiratory Medicine, The First Hospital of Jilin University Changchun 130021 China
- Key Laboratory of Luminescence Science and Technology, Chinese Academy of Sciences & State Key Laboratory of Luminescence and Applications, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences Changchun 130033 Jilin China
| | - Hongsen Liu
- Key Laboratory of Luminescence Science and Technology, Chinese Academy of Sciences & State Key Laboratory of Luminescence and Applications, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences Changchun 130033 Jilin China
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province Hangzhou 310000 China
| | - Yanfei Zhao
- Department of Respiratory Medicine, The First Hospital of Jilin University Changchun 130021 China
| | - Haoran Chen
- Key Laboratory of Luminescence Science and Technology, Chinese Academy of Sciences & State Key Laboratory of Luminescence and Applications, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences Changchun 130033 Jilin China
| | - Qiqing Li
- Key Laboratory of Luminescence Science and Technology, Chinese Academy of Sciences & State Key Laboratory of Luminescence and Applications, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences Changchun 130033 Jilin China
| | - Xiaodan Li
- Department of Respiratory Medicine, The First Hospital of Jilin University Changchun 130021 China
| | - Shucheng Hua
- Department of Respiratory Medicine, The First Hospital of Jilin University Changchun 130021 China
| | - Dianbo Cao
- Department of Radiology, The First Hospital of Jilin University Changchun 130021 China
| | - Yulei Chang
- Key Laboratory of Luminescence Science and Technology, Chinese Academy of Sciences & State Key Laboratory of Luminescence and Applications, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences Changchun 130033 Jilin China
| |
Collapse
|
2
|
Nkune NW, Abrahamse H. The phototoxic effect of a gold-antibody-based nanocarrier of phthalocyanine on melanoma monolayers and tumour spheroids. RSC Adv 2024; 14:19490-19504. [PMID: 38895533 PMCID: PMC11184583 DOI: 10.1039/d4ra03858d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 06/11/2024] [Indexed: 06/21/2024] Open
Abstract
In recent years, photodynamic therapy (PDT) has garnered significant attention in cancer treatment due to its increased potency and non-invasiveness compared to conventional therapies. Active-targeted delivery of photosensitizers (PSs) is a mainstay strategy to significantly reduce its off-target toxicity and enhance its phototoxic efficacy. The anti-melanoma inhibitory activity (MIA) antibody is a targeting biomolecule that can be integrated into a nanocarrier system to actively target melanoma cells due to its specific binding to MIA antigens that are highly expressed on the surface of melanoma cells. Gold nanoparticles (AuNPs) are excellent nanocarriers due to their ability to encapsulate a variety of therapeutics, such as PSs, and their ability to bind with targeting moieties for improved bioavailability in cancer cells. Hence, we designed a nanobioconjugate (NBC) composed of zinc phthalocyanine tetrasulfonic acid (ZnPcS4), AuNPs and anti-MIA Ab to improve ZnPcS4 bioavailability and phototoxicity in two and three-dimensional tumour models. In summary, we demonstrated that this nanobioconjugate showed significant inhibitory effects on both melanoma models due to increased ROS yields and bioavailability of the melanoma cells compared to free ZnPcS4.
Collapse
Affiliation(s)
- Nkune Williams Nkune
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg P.O. Box 17011 Doornfontein 2028 South Africa +27-11-559-655
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg P.O. Box 17011 Doornfontein 2028 South Africa +27-11-559-655
| |
Collapse
|
3
|
Nkune NW, Abrahamse H. The Efficacy of Zinc Phthalocyanine Nanoconjugate on Melanoma Cells Grown as Three-Dimensional Multicellular Tumour Spheroids. Pharmaceutics 2023; 15:2264. [PMID: 37765232 PMCID: PMC10535874 DOI: 10.3390/pharmaceutics15092264] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Melanoma remains a major public health concern that is highly resistant to standard therapeutic approaches. Photodynamic therapy (PDT) is an underutilised cancer therapy with an increased potency and negligible side effects, and it is non-invasive compared to traditional treatment modalities. Three-dimensional multicellular tumour spheroids (MCTS) closely resemble in vivo avascular tumour features, allowing for the more efficient and precise screening of novel anticancer agents with various treatment combinations. In this study, we utilised A375 human melanoma spheroids to screen the phototoxic effect of zinc phthalocyanine tetrasulfonate (ZnPcS4) conjugated to gold nanoparticles (AuNP). The nanoconjugate was synthesised and characterised using ultraviolet-visible spectroscopy, a high-resolution transmission electron microscope (TEM), dynamic light scattering (DLS), and zeta potential (ZP). The phototoxicity of the nanoconjugate was tested on the A375 MCTS using PDT at a fluency of 10 J/cm2. After 24 h, the cellular responses were evaluated via microscopy, an MTT viability assay, an ATP luminescence assay, and cell death induction using annexin propidium iodide. The MTT viability assay demonstrated that the photoactivated ZnPcS4, at a concentration of 12.73 µM, caused an approximately 50% reduction in the cell viability of the spheroids. When conjugated to AuNPs, the latter significantly increased the cellular uptake and cytotoxicity in the melanoma spheroids via the induction of apoptosis. This novel Zinc Phthalocyanine Nanoconjugate shows promise as a more effective PDT treatment modality.
Collapse
Affiliation(s)
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, P.O. Box 17011, Johannesburg 2028, South Africa;
| |
Collapse
|
4
|
Ma C, Wang Y, Chen W, Hou T, Zhang H, Zhang H, Yao X, Xia C. Caspase-1 Regulates the Apoptosis and Pyroptosis Induced by Phthalocyanine Zinc-Mediated Photodynamic Therapy in Breast Cancer MCF-7 Cells. Molecules 2023; 28:5934. [PMID: 37630186 PMCID: PMC10458486 DOI: 10.3390/molecules28165934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/29/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
Photodynamic therapy (PDT) is an innovative and perspective antineoplastic therapy. Tetra-α-(4-carboxyphenoxy) phthalocyanine zinc (TαPcZn)-mediated PDT (TαPcZn-PDT) has shown antitumor activity in some tumor cells, but the manner in which caspase-1 is involved in the regulation of apoptosis and pyroptosis in the TαPcZn-PDT-treated breast cancer MCF-7 cells is unclear. Therefore, effects of TαPcZn-PDT on cytotoxicity, cell viability, apoptosis, pyroptosis, cellular reactive oxygen species (ROS), mitochondrial membrane potential (ΔΨm), caspase-1, caspase-3, and nuclear transcription factor-κB (NFκB) in MCF-7 cells was firstly examined in the present study. The findings demonstrated that TαPcZn-PDT resulted in the increase in cytotoxicity and the percentage of apoptotic and pyroptotic cells, the reduction in cell viability and ΔΨm, the production of ROS and the activation of caspase-1, caspase-3 and NFκB in MCF-7 cells. Furthermore, the results also revealed that siRNA-targeting caspase-1 (siRNA-caspase-1) attenuated the effect of TαPcZn-PDT on apoptosis, pyroptosis and the activation of caspase-1, caspase-3 and NFκB in MCF-7 cells. Taken together, we conclude that caspase-1 regulates the apoptosis and pyroptosis induced by TαPcZn-PDT in MCF-7 cells.
Collapse
Affiliation(s)
- Chunjie Ma
- Pharmacy Department, Qiqihar Medical University, Qiqihar 161006, China; (C.M.); (T.H.); (H.Z.); (H.Z.); (X.Y.)
| | - Yu Wang
- Basic Medicine Department, Qiqihar Medical University, Qiqihar 161006, China;
| | - Wei Chen
- College of Chemistry and Chemical Engineering, Qiqihar University, Qiqihar 161000, China;
| | - Ting Hou
- Pharmacy Department, Qiqihar Medical University, Qiqihar 161006, China; (C.M.); (T.H.); (H.Z.); (H.Z.); (X.Y.)
| | - Honglian Zhang
- Pharmacy Department, Qiqihar Medical University, Qiqihar 161006, China; (C.M.); (T.H.); (H.Z.); (H.Z.); (X.Y.)
| | - Hongguang Zhang
- Pharmacy Department, Qiqihar Medical University, Qiqihar 161006, China; (C.M.); (T.H.); (H.Z.); (H.Z.); (X.Y.)
| | - Xu Yao
- Pharmacy Department, Qiqihar Medical University, Qiqihar 161006, China; (C.M.); (T.H.); (H.Z.); (H.Z.); (X.Y.)
| | - Chunhui Xia
- Pharmacy Department, Qiqihar Medical University, Qiqihar 161006, China; (C.M.); (T.H.); (H.Z.); (H.Z.); (X.Y.)
| |
Collapse
|
5
|
Lima E, Reis LV. Photodynamic Therapy: From the Basics to the Current Progress of N-Heterocyclic-Bearing Dyes as Effective Photosensitizers. Molecules 2023; 28:5092. [PMID: 37446758 DOI: 10.3390/molecules28135092] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/16/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Photodynamic therapy, an alternative that has gained weight and popularity compared to current conventional therapies in the treatment of cancer, is a minimally invasive therapeutic strategy that generally results from the simultaneous action of three factors: a molecule with high sensitivity to light, the photosensitizer, molecular oxygen in the triplet state, and light energy. There is much to be said about each of these three elements; however, the efficacy of the photosensitizer is the most determining factor for the success of this therapeutic modality. Porphyrins, chlorins, phthalocyanines, boron-dipyrromethenes, and cyanines are some of the N-heterocycle-bearing dyes' classes with high biological promise. In this review, a concise approach is taken to these and other families of potential photosensitizers and the molecular modifications that have recently appeared in the literature within the scope of their photodynamic application, as well as how these compounds and their formulations may eventually overcome the deficiencies of the molecules currently clinically used and revolutionize the therapies to eradicate or delay the growth of tumor cells.
Collapse
Affiliation(s)
- Eurico Lima
- CQ-VR-Chemistry Centre of Vila Real, University of Trás-os-Montes and Alto Douro, Quinta de Prados, 5001-801 Vila Real, Portugal
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6201-506 Covilhã, Portugal
| | - Lucinda V Reis
- CQ-VR-Chemistry Centre of Vila Real, University of Trás-os-Montes and Alto Douro, Quinta de Prados, 5001-801 Vila Real, Portugal
| |
Collapse
|
6
|
Chilakamarthi U, Mahadik NS, Koteshwar D, Krishna NV, Giribabu L, Banerjee R. Potentiation of novel porphyrin based photodynamic therapy against colon cancer with low dose doxorubicin and elucidating the molecular signalling pathways responsible for relapse. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 238:112625. [PMID: 36529058 DOI: 10.1016/j.jphotobiol.2022.112625] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/06/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Photodynamic therapy (PDT) is a promising non-invasive treatment modality for cancer and can be potentiated by combination with chemotherapy. Here, we combined PDT of novel porphyrin-based photosensitizers with low dose doxorubicin (Dox) to get maximum outcome. Dox potentiated and showed synergism with PDT under in vitro conditions on CT26.WT cells. The current colon cancer treatment strategies assure partial or even complete tumour regression but loco-regional relapse or distant metastasis is the major cause of death despite combination therapy. The spared cells after the treatment contribute to relapse and it is important to study their behaviour in host environment. Hence, we developed relapse models for PDT, Dox and combination treatments by transplanting respectively treated equal number of live cells to mice (n = 5) for tumour formation. Most of the treated cells lost tumour forming ability, but some treatment resistant cells developed tumours in few mice. These tumours served as relapse models and Western blot analysis of tumour samples provided clinically relevant information to delineate resistance strategies of individual as well as combination therapies at molecular level. Our results showed that low dose Dox helped in increasing the tumour inhibiting effect of PDT in combination therapy, but still there are indeed possibilities of relapse at later stages due to chemoresistance and immune suppression that may occur post-treatment. We observed that the combination therapy may also lead to the development of multidrug resistant (MDR) phenotype during relapse. Thus, this study provided clinically relevant information to further strengthen and improve PDT-drug combination therapy in order to avoid relapse and to treat cancer more effectively.
Collapse
Affiliation(s)
- Ushasri Chilakamarthi
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India.
| | - Namita S Mahadik
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Devulapally Koteshwar
- Polymers and Functional Materials Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Narra Vamsi Krishna
- Polymers and Functional Materials Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Lingamallu Giribabu
- Polymers and Functional Materials Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India.
| | - Rajkumar Banerjee
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India.
| |
Collapse
|
7
|
Vazirabad AF, Noorolyai S, Baghbani E, Mahboob S, Zargari F, Rahmani S, Sorkhabi A, Montazami N, Sameti P, Baradaran B. Silencing of SiX-4 enhances the chemosensitivity of melanoma cells to Cisplatin. Pathol Res Pract 2022; 240:154194. [PMID: 36370483 DOI: 10.1016/j.prp.2022.154194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 10/23/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
Abstract
Melanoma is the riskiest type of skin cancer. Its prevalence has been rapidly increased over the last three decades. SIX1, SIX2, SIX3, SIX4, SIX5, and SIX6 are members of the sine oculis homeobox (SIX) homolog family. It is imperative to identify new melanoma biomarkers to improve the predictive value for melanoma prognosis, which could enhance our understanding of carcinogenesis and tumor progression. In this study, we investigated whether silencing of SIX4 in a melanoma cell line (A375 cells) in combination with Cisplatin can affect the apoptosis and suppression of cell cycle progression, migration of the melanoma cells. MTT test and colony formation assay was applied to determine the IC50 of Cisplatin and the combined effect of SIX4 siRNA and Cisplatin on the viability and clonogenesis of the A-375 cells. qRT-PCR was performed to determine the c-myc, BCL-2, BAX, MMP-9, CXCR4, and Rock genes expression. Furthermore, flow cytometry was applied to evaluate apoptosis, autophagy, and the cell cycle status in different groups. Finally, wound healing assay was employed to evaluate the effect of this combination therapy on migratory capacity. SIX4 suppression increased the chemosensitivity of A-375 cells to Cisplatin and decreased its efficient dose. Furthermore, SIX4 suppression alongside Cisplatin reduced cell migration rate, arrested the cell cycle at the G1 phase, induced apoptosis by modulating the expression of apoptotic target genes, induced autophagy, and also significantly inhibits clonogenesis of A-375 cells. SIX4 plays a significant role in the chemosensitivity and pathogenesis of melanoma. Therefore, SIX4 suppression, in combination with Cisplatin, may be a promising therapeutic approach in treating melanoma.
Collapse
Affiliation(s)
| | - Saeed Noorolyai
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soltanali Mahboob
- Faculty of Health and Nutrition, Tabriz University of Medical Sciences, Department of Food and Nutrition Security, Iran; Department of Biochemistry, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Felor Zargari
- Department of Medical Science, Marand Branch, Islamic Azad University, Marand, Iran
| | - Shima Rahmani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Sorkhabi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nooshin Montazami
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pouriya Sameti
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
8
|
Rajabi N, Mohammadnejad F, Doustvandi MA, Shadbad MA, Amini M, Tajalli H, Mokhtarzadeh A, Baghbani E, Silvestris N, Baradaran B. Photodynamic therapy with zinc phthalocyanine enhances the anti-cancer effect of tamoxifen in breast cancer cell line: Promising combination treatment against triple-negative breast cancer? Photodiagnosis Photodyn Ther 2022; 41:103212. [PMID: 36436735 DOI: 10.1016/j.pdpdt.2022.103212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 11/12/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022]
Abstract
Photodynamic therapy (PDT) is a light-based anti-neoplastic therapeutic approach. Growing evidence indicates that combining conventional anti-cancer therapies with PDT can be a promising approach to treat malignancies. Herein, we aimed to investigate anti-cancer effects of the combination treatment of zinc phthalocyanine (ZnPc)-PDT with tamoxifen (TA) on MDA-MB-231 cells (as a triple-negative breast cancer (TNBC) cell line). For this purpose, we investigated the cytotoxicity of TA and ZnPc-PDT on MDA-MB-231 cells performing the MTT assay. The effect of TA and ZnPc-PDT on the apoptosis of MDA-MB-231 cells was studied using Annexin V/PI and DAPI staining. The wound-healing assay, and colony formation assay were performed to study the effect of TA and ZnPc-PDT on the migration, and clonogenicity of MDA-MB-231 cells, respectively. The qRT-PCR was done to study the gene expression of caspase-8, caspase-9, caspase-3, ZEB1, ROCK1, SNAIL1, CD133, CD44, SOX2, and ABCG2 (ATP-binding cassette sub-family G member 2). Based on our results, monotherapies with TA and ZnPc-PDT can remarkably increase cell cytotoxicity effects, stimulate apoptosis via downregulating Bcl-2 and upregulating caspase-3 and caspase-9, inhibit migration via downregulating SNAIL1 and ZEB1, and suppress clonogenicity via downregulating SOX2 and CD44 in MDA-MB-231 cells. Besides, these monotherapies can downregulate the expression of ABCG2 in MDA-MB-231 cells. Nevertheless, the combination treatment can potentiate the above-mentioned anti-cancer effects compared to monotherapy with TA. Of interest, the combined treatment of TA with ZnPc-PDT can synergically increase cell cytotoxicity effects on MDA-MB-231 cells. In fact, synergistic effects were estimated by calculation of Combination Index (CI); that synergistic outcomes were observed in all groups. Also, this combination treatment can significantly upregulate the caspase-8 gene expression and downregulate ROCK1 and CD133 gene expression in MDA-MB-231 cells. Overall, our results show that ZnPc-PDT can more sensitize the MDA-MB-231 cells to TA treatment. Based on our knowledge and experiment, the synergistic effects of ZnPc-PDT and TA deserve further evaluation in cancer research.
Collapse
Affiliation(s)
- Neda Rajabi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Amin Doustvandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Habib Tajalli
- Biophotonic Research Center, Islamic Azad University, Tabriz Branch, Tabriz, Iran; Research Institute for Applied Physics and Astronomy, University of Tabriz, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nicola Silvestris
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina, Italy.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
9
|
Malindi Z, Barth S, Abrahamse H. The Potential of Antibody Technology and Silver Nanoparticles for Enhancing Photodynamic Therapy for Melanoma. Biomedicines 2022; 10:2158. [PMID: 36140259 PMCID: PMC9495799 DOI: 10.3390/biomedicines10092158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022] Open
Abstract
Melanoma is highly aggressive and is known to be efficient at resisting drug-induced apoptotic signals. Resection is currently the gold standard for melanoma management, but it only offers local control of the early stage of the disease. Metastatic melanoma is prone to recurrence, and has a poor prognosis and treatment response. Thus, the need for advanced theranostic alternatives is evident. Photodynamic therapy has been increasingly studied for melanoma treatment; however, it relies on passive drug accumulation, leading to off-target effects. Nanoparticles enhance drug biodistribution, uptake and intra-tumoural concentration and can be functionalised with monoclonal antibodies that offer selective biorecognition. Antibody-drug conjugates reduce passive drug accumulation and off-target effects. Nonetheless, one limitation of monoclonal antibodies and antibody-drug conjugates is their lack of versatility, given cancer's heterogeneity. Monoclonal antibodies suffer several additional limitations that make recombinant antibody fragments more desirable. SNAP-tag is a modified version of the human DNA-repair enzyme, O6-alkylguanine-DNA alkyltransferase. It reacts in an autocatalytic and covalent manner with benzylguanine-modified substrates, providing a simple protein labelling system. SNAP-tag can be genetically fused with antibody fragments, creating fusion proteins that can be easily labelled with benzylguanine-modified payloads for site-directed delivery. This review aims to highlight the benefits and limitations of the abovementioned approaches and to outline how their combination could enhance photodynamic therapy for melanoma.
Collapse
Affiliation(s)
- Zaria Malindi
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, 55 Beit Street, Doornfontein, Johannesburg 2028, South Africa
| | - Stefan Barth
- Medical Biotechnology and Immunotherapy Research Unit, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road Observatory, Cape Town 7925, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, 55 Beit Street, Doornfontein, Johannesburg 2028, South Africa
| |
Collapse
|
10
|
Advantages of combined photodynamic therapy in the treatment of oncological diseases. Biophys Rev 2022; 14:941-963. [DOI: 10.1007/s12551-022-00962-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/24/2022] [Indexed: 12/22/2022] Open
|
11
|
Atta-Eyison AA, Anukwah GD, Zugle R. Photocatalysis using zinc oxide-zinc phthalocyanine composite for effective mineralization of organic pollutants. CATAL COMMUN 2021. [DOI: 10.1016/j.catcom.2021.106357] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
12
|
Catalani E, Giovarelli M, Zecchini S, Perrotta C, Cervia D. Oxidative Stress and Autophagy as Key Targets in Melanoma Cell Fate. Cancers (Basel) 2021; 13:cancers13225791. [PMID: 34830947 PMCID: PMC8616245 DOI: 10.3390/cancers13225791] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/10/2021] [Accepted: 11/17/2021] [Indexed: 01/18/2023] Open
Abstract
Melanoma originates from the malignant transformation of melanocytes and is one of the most aggressive forms of cancer. The recent approval of several drugs has increased the chance of survival although a significant subset of patients with metastatic melanoma do not show a long-lasting response to these treatments. The complex cross-talk between oxidative stress and the catabolic process autophagy seems to play a central role in all aspects of melanoma pathophysiology, from initiation to progression and metastasis, including drug resistance. However, determining the fine role of autophagy in cancer death and in response to redox disruption is still a fundamental challenge in order to advance both basic and translational aspects of this field. In order to summarize the interactions among reactive oxygen and nitrogen species, autophagy machinery and proliferation/growth/death/apoptosis/survival, we provide here a narrative review of the preclinical evidence for drugs/treatments that modulate oxidative stress and autophagy in melanoma cells. The significance and the potential for pharmacological targeting (also through multiple and combination approaches) of these two different events, which can contribute independently or simultaneously to the fate of melanoma, may help to define new processes and their interconnections underlying skin cancer biology and unravel new reliable approaches.
Collapse
Affiliation(s)
- Elisabetta Catalani
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, Largo dell’Università snc, 01100 Viterbo, Italy;
| | - Matteo Giovarelli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, Via G.B. Grassi 74, 20157 Milano, Italy; (M.G.); (S.Z.)
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, Via G.B. Grassi 74, 20157 Milano, Italy; (M.G.); (S.Z.)
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, Via G.B. Grassi 74, 20157 Milano, Italy; (M.G.); (S.Z.)
- Correspondence: (C.P.); (D.C.)
| | - Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, Largo dell’Università snc, 01100 Viterbo, Italy;
- Correspondence: (C.P.); (D.C.)
| |
Collapse
|
13
|
Gholizadeh M, Doustvandi MA, Mohammadnejad F, Shadbad MA, Tajalli H, Brunetti O, Argentiero A, Silvestris N, Baradaran B. Photodynamic Therapy with Zinc Phthalocyanine Inhibits the Stemness and Development of Colorectal Cancer: Time to Overcome the Challenging Barriers? Molecules 2021; 26:6877. [PMID: 34833970 PMCID: PMC8621355 DOI: 10.3390/molecules26226877] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 02/07/2023] Open
Abstract
Photodynamic therapy (PDT) is a light-based cancer therapy approach that has shown promising results in treating various malignancies. Growing evidence indicates that cancer stem cells (CSCs) are implicated in tumor recurrence, metastasis, and cancer therapy resistance in colorectal cancer (CRC); thus, targeting these cells can ameliorate the prognosis of affected patients. Based on our bioinformatics results, SOX2 overexpression is significantly associated with inferior disease-specific survival and worsened the progression-free interval of CRC patients. Our results demonstrate that zinc phthalocyanine (ZnPc)-PDT with 12 J/cm2 or 24 J/cm2 irradiation can substantially decrease tumor migration via downregulating MMP9 and ROCK1 and inhibit the clonogenicity of SW480 cells via downregulating CD44 and SOX2. Despite inhibiting clonogenicity, ZnPc-PDT with 12 J/cm2 irradiation fails to downregulate CD44 expression in SW480 cells. Our results indicate that ZnPc-PDT with 12 J/cm2 or 24 J/cm2 irradiation can substantially reduce the cell viability of SW480 cells and stimulate autophagy in the tumoral cells. Moreover, our results show that ZnPc-PDT with 12 J/cm2 or 24 J/cm2 irradiation can substantially arrest the cell cycle at the sub-G1 level, stimulate the intrinsic apoptosis pathway via upregulating caspase-3 and caspase-9 and downregulating Bcl-2. Indeed, our bioinformatics results show considerable interactions between the studied CSC-related genes with the studied migration- and apoptosis-related genes. Collectively, the current study highlights the potential role of ZnPc-PDT in inhibiting stemness and CRC development, which can ameliorate the prognosis of CRC patients.
Collapse
Affiliation(s)
- Mahsa Gholizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51666-14731, Iran; (M.G.); (M.A.D.); (F.M.); (M.A.S.)
| | - Mohammad Amin Doustvandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51666-14731, Iran; (M.G.); (M.A.D.); (F.M.); (M.A.S.)
| | - Fateme Mohammadnejad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51666-14731, Iran; (M.G.); (M.A.D.); (F.M.); (M.A.S.)
| | - Mahdi Abdoli Shadbad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51666-14731, Iran; (M.G.); (M.A.D.); (F.M.); (M.A.S.)
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 51666-14731, Iran
| | - Habib Tajalli
- Biophotonic Research Center, Islamic Azad University, Tabriz Branch, Tabriz 51579-44533, Iran;
- Research Institute for Applied Physics and Astronomy, University of Tabriz, Tabriz 51666-16471, Iran
| | - Oronzo Brunetti
- Istituto Tumori BariGiovanni Paolo II, Istituto Nazionale dei Tumori (IRCCS), 70124 Bari, Italy; (O.B.); (A.A.)
| | - Antonella Argentiero
- Istituto Tumori BariGiovanni Paolo II, Istituto Nazionale dei Tumori (IRCCS), 70124 Bari, Italy; (O.B.); (A.A.)
| | - Nicola Silvestris
- Istituto Tumori BariGiovanni Paolo II, Istituto Nazionale dei Tumori (IRCCS), 70124 Bari, Italy; (O.B.); (A.A.)
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51666-14731, Iran; (M.G.); (M.A.D.); (F.M.); (M.A.S.)
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz 51666-14731, Iran
| |
Collapse
|
14
|
Martínez-Edo G, Xue EY, Ha SYY, Pontón I, González-Delgado JA, Borrós S, Torres T, Ng DKP, Sánchez-García D. Nanoparticles for Triple Drug Release for Combined Chemo- and Photodynamic Therapy. Chemistry 2021; 27:14610-14618. [PMID: 34460988 DOI: 10.1002/chem.202101842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Indexed: 12/13/2022]
Abstract
A pH-responsive drug delivery system (DDS) based on mesoporous silica nanoparticles (MSNs) has been prepared for the delivery of three anticancer drugs with different modes of action. The novelty of this system is its ability to combine synergistic chemotherapy and photodynamic therapy. A photoactive conjugate of a phthalocyanine (Pc) and a topoisomerase I inhibitor (topo-I), namely camptothecin (CPT), linked by a poly(ethylene glycol) (PEG) chain has been synthesized and then loaded into the mesopores of MSNs. Doxorubicin (DOX), which is a topoisomerase II inhibitor (topo-II), has also been covalently anchored to the outer surface of the MSNs through a dihydrazide PEG linker. In the acidic environment of tumor cells, selective release of the three drugs takes place. In vitro studies have demonstrated the endocytosis of the system into HeLa and HepG2 cells, and the subsequent release of the three drugs into the cytoplasm and nucleus. Furthermore, the cytotoxic effect of DOX, CPT and Pc has been assessed in vitro before and upon light irradiation.
Collapse
Affiliation(s)
- Gabriel Martínez-Edo
- Grup d'Enginyera de Materials (GEMAT), Institut Químic de Sarrià, Universitat Ramon Llull, Via Augusta 390, 08017, Barcelona, Spain
| | - Evelyn Y Xue
- Department of Chemistry, The Chinese University of Hong Kong Shatin, N.T., Hong Kong, China
| | - Summer Y Y Ha
- Department of Chemistry, The Chinese University of Hong Kong Shatin, N.T., Hong Kong, China
| | - Iris Pontón
- Grup d'Enginyera de Materials (GEMAT), Institut Químic de Sarrià, Universitat Ramon Llull, Via Augusta 390, 08017, Barcelona, Spain
| | - José Antonio González-Delgado
- Department of Organic Chemistry and Institute for Advanced Research in Chemistry (IAdChem), Universidad Autónoma de Madrid, c/ Francisco Tomás y Valiente 7 Cantoblanco, 28049, Madrid, Spain
| | - Salvador Borrós
- Grup d'Enginyera de Materials (GEMAT), Institut Químic de Sarrià, Universitat Ramon Llull, Via Augusta 390, 08017, Barcelona, Spain
| | - Tomás Torres
- Department of Organic Chemistry and Institute for Advanced Research in Chemistry (IAdChem), Universidad Autónoma de Madrid, c/ Francisco Tomás y Valiente 7 Cantoblanco, 28049, Madrid, Spain.,IMDEA-Nanociencia, c/ Faraday 9, Campus de Cantoblanco, 28049, Madrid, Spain
| | - Dennis K P Ng
- Department of Chemistry, The Chinese University of Hong Kong Shatin, N.T., Hong Kong, China
| | - David Sánchez-García
- Grup d'Enginyera de Materials (GEMAT), Institut Químic de Sarrià, Universitat Ramon Llull, Via Augusta 390, 08017, Barcelona, Spain
| |
Collapse
|
15
|
Moghassemi S, Dadashzadeh A, de Souza PEN, Azevedo RB, Amorim CA. AlPc/ZnPc-based oncological photodynamic therapy for a selective eradication of leukemic cells from ovarian tissue. Photodiagnosis Photodyn Ther 2021; 36:102555. [PMID: 34597832 DOI: 10.1016/j.pdpdt.2021.102555] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/30/2021] [Accepted: 09/24/2021] [Indexed: 01/10/2023]
Abstract
Due to the risk of reintroducing malignant cells, autotransplantation of cryopreserved ovarian tissue is not allowed in leukemia patients. In order to restore fertility in these patients, ex vivo purging of ovarian fragments could be proposed as a strategy to eradicate malignant cells before grafting. Photodynamic therapy (PDT), as a clinical-approved modality, is a minimally invasive and selective therapeutic for eliminating malignant cells. The present work aims therefore to evaluate the phototoxicity of two photosensitizers (aluminum phthalocyanine (AlPc) and zinc phthalocyanine (ZnPc)) on leukemia cells. To this end, two lines of malignant cells (K562 and HL-60) and isolated ovarian stromal cells (control) were treated by PDT using a diode laser with various energy densities. Cell viability after the treatment, the amount of generated reactive oxygen species, dark toxicity of the photosensitizers, and single-cell morphology were studied. Our results demonstrated that using irradiation with the energy density of 10 J/cm2, 1 µM AlPc could significantly reduce the viability of K562 (4.73 ± 0.14%) and HL-60 (2.74 ± 0.31%). Similarly, the viability of these cells was reduced (K562 cells: 3.84 ± 0.81%; HL-60 cells: 6.82 ± 3.21%) with 1 µM ZnPc and an energy density of 50 J/cm2. On the other hand, these PDT protocols had no significant effect on stromal cells. These findings indicate that our approach can be a promising strategy for the safe restoration of fertility in leukemia patients. However, further studies are necessary to assess its efficiency in ovarian fragments containing malignant cells to determine their eradication rate and the effect of our treatment on the survival of stromal cells and preantral follicles.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | | | - Ricardo Bentes Azevedo
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília, Brasília DF, Brazil
| | - Christiani A Amorim
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
16
|
Solğun DG, Yıldıko Ü, Ağırtaş MS. Synthesis, DFT Calculations, Photophysical, Photochemical Properties of Peripherally Metallophthalocyanines Bearing (2-(Benzo[d] [1,3] Dioxol-5-Ylmethoxy) Phenoxy) Substituents. Polycycl Aromat Compd 2021. [DOI: 10.1080/10406638.2021.1983618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Derya Güngördü Solğun
- Department of Chemistry, Faculty of Science, Van Yuzuncu Yıl University, Van, Turkey
| | - Ümit Yıldıko
- Architecture and Engineering Faculty, Department of Bioengineering, Kafkas University, Kars, Turkey
| | - Mehmet Salih Ağırtaş
- Department of Chemistry, Faculty of Science, Van Yuzuncu Yıl University, Van, Turkey
| |
Collapse
|
17
|
Zeinali S, Tuncel A, Yüzer A, Yurt F. Imaging and detection of cell apoptosis byIn vitrophotodynamic therapy applications of zinc (II) phthalocyanine on human melanoma cancer. Photodiagnosis Photodyn Ther 2021; 36:102518. [PMID: 34478898 DOI: 10.1016/j.pdpdt.2021.102518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/29/2021] [Accepted: 08/27/2021] [Indexed: 01/10/2023]
Abstract
This study aims to investigate the photodynamic therapy (PDT) effects on MeWo (human melanoma cells) and HaCaT (normal human keratinocyte cells) by light stimulation of different concentrations of Zinc (II)-tetra-tert-butyl-phthalocyaninato (ZnPc). MTT viability assay data indicated that a 25 μM concentration of ZnPc is cytotoxic to the melanoma cancer cells while this concentration of ZnPc is not cytotoxic for the HaCaT cell line. Moreover, the results showed that photoactivated ZnPc at 12.5 μM concentration reduced the cell viability of the MeWo cell line to about 50 %. At this photosensitizing concentration, the efficacy of light doses of 20, 30, 40, and 50 J/cm2 was evaluated against MeWo and HaCaT cells. ZnPc at a concentration of 12.5 μM activated with a light dose of 50 J/cm2 was the most efficient for the killing of MeWo cells. In conclusion, the 12.5 μM of ZnPc with the treatment light dose of 50 J/cm2 from a RED light source was adequate to destroy MeWo cells by the ROS-induced apoptosis mechanism. It also exhibited low killing effects on healthy HaCaT cells. These findings are supported by the results of apoptosis with the Annexin V & Dead Cell Kit and fluorescence imaging.
Collapse
Affiliation(s)
- Sevda Zeinali
- Department Biomedical Technologies, Institute of Science, Ege University, 35100, Bornova, Izmir, Turkey
| | - Ayca Tuncel
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, 35100, Bornova, Izmir, Turkey
| | - Abdulcelil Yüzer
- Faculty of Engineering, Department of Engineering Fundamental Sciences, Tarsus University, 33400, Tarsus, Turkey
| | - Fatma Yurt
- Department Biomedical Technologies, Institute of Science, Ege University, 35100, Bornova, Izmir, Turkey; Department of Nuclear Applications, Institute of Nuclear Science, Ege University, 35100, Bornova, Izmir, Turkey.
| |
Collapse
|
18
|
Co-Encapsulation of Methylene Blue and PARP-Inhibitor into Poly(Lactic-Co-Glycolic Acid) Nanoparticles for Enhanced PDT of Cancer. NANOMATERIALS 2021; 11:nano11061514. [PMID: 34201069 PMCID: PMC8227603 DOI: 10.3390/nano11061514] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/16/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022]
Abstract
The development of resistance against photodamage triggered by photodynamic therapy (PDT) is ascribed mainly to the cellular redox defenses and repair. If the tumor tissue is not promptly eliminated by the first few PDT sessions, PDT-resistance can be favored, challenging the efficacy of the treatment. Although the mechanism of PDT resistance is still unclear, in vitro assays have evidenced that it can be developed through the PARP damage-repair signaling pathway. Therefore, inhibition of poly(adenosine diphosphate (ADP)-ribose) polymerase (PARP) has the potential to increase PDT efficacy. This work reports on the synthesis of a controlled release system of a photosensitizer, methylene blue (MB) and a PARP-inhibitor, the veliparib. MB and veliparib were co-encapsulated in poly(lactic-co-glycolic acid) (PLGA) nanoparticles (VMB-NPs). A colloidal stable aqueous suspension of nanoparticles was obtained. The average hydrodynamic diameter was 90 nm and a narrow size distribution was obtained, with a polydispersity index (PDI) of 0.08. The release kinetics of MB and veliparib from VMB-NPs showed an initial burst of 8.7% and 58.3% release of the total amounts of MB and veliparib respectively, in the first 6 h, and a delayed release of up to 11.3% and 70%, in 19 days, for MB and veliparib, respectively. The VMB-NPs showed no cytotoxicity in the dark but the viability of B16F10-Nex2 cells decreased by 36% when the cells were irradiated (102 J/cm2, 660 nm) and treated with VMB-NPs containing 1.0 µM of MB and 8.3 µM of veliparib. Considering the increased photoactivity even at low MB and veliparib concentrations and the absence of cytotoxicity in dark, the co-encapsulation of MB and veliparib was shown to be a promising strategy to improve the PDT efficacy.
Collapse
|
19
|
Lange N, Szlasa W, Saczko J, Chwiłkowska A. Potential of Cyanine Derived Dyes in Photodynamic Therapy. Pharmaceutics 2021; 13:818. [PMID: 34072719 PMCID: PMC8229084 DOI: 10.3390/pharmaceutics13060818] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/28/2021] [Accepted: 05/28/2021] [Indexed: 12/26/2022] Open
Abstract
Photodynamic therapy (PDT) is a method of cancer treatment that leads to the disintegration of cancer cells and has developed significantly in recent years. The clinically used photosensitizers are primarily porphyrin, which absorbs light in the red spectrum and their absorbance maxima are relatively short. This review presents group of compounds and their derivatives that are considered to be potential photosensitizers in PDT. Cyanine dyes are compounds that typically absorb light in the visible to near-infrared-I (NIR-I) spectrum range (750-900 nm). This meta-analysis comprises the current studies on cyanine dye derivatives, such as indocyanine green (so far used solely as a diagnostic agent), heptamethine and pentamethine dyes, squaraine dyes, merocyanines and phthalocyanines. The wide array of the cyanine derivatives arises from their structural modifications (e.g., halogenation, incorporation of metal atoms or organic structures, or synthesis of lactosomes, emulsions or conjugation). All the following modifications aim to increase solubility in aqueous media, enhance phototoxicity, and decrease photobleaching. In addition, the changes introduce new features like pH-sensitivity. The cyanine dyes involved in photodynamic reactions could be incorporated into sets of PDT agents.
Collapse
Affiliation(s)
- Natalia Lange
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland; (N.L.); (W.S.)
| | - Wojciech Szlasa
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland; (N.L.); (W.S.)
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| | - Agnieszka Chwiłkowska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| |
Collapse
|
20
|
Yousefi Sadeghloo A, Khorsandi K, Kianmehr Z. Synergistic effect of photodynamic treatment and doxorubicin on triple negative breast cancer cells. Photochem Photobiol Sci 2021; 19:1580-1589. [PMID: 33030191 DOI: 10.1039/d0pp00132e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Breast cancer is a metastatic cancer that can spread to other organs, such as the bone, liver, and brain. There are many treatments for breast cancer, such as surgery and chemotherapy, but they lead to resistance and side effects. Therefore, the discovery of new therapies with high efficacy and low toxicity that selectively affect cancer cells is of great importance. Of late, the combination therapy has been suggested as a novel approach compared to existing treatments. In the present study, the effect of the combined treatment of doxorubicin (DOX) and methylene blue activated in the presence of laser irradiation (PDT) on triple-negative breast cancer cells has been investigated. Human breast cancer cell line MDA-MB-231 was exposed to different concentrations of DOX, methylene blue (MB) and DOX-methylene blue (MB-DOX) combination therapy in two different conditions: first the treatment with DOX and then with MB-PDT, and another treatment first with MB-PDT and then with DOX. Cell viability was evaluated using the MTT assay. Morphological and colonization changes were observed by light microscopy. The occurrence of apoptotic cell death was assessed by double-staining ethidium bromide-acridine orange using fluorescence microscopy and flow cytometry. The results showed that the combination of using MB-PDT, followed by DOX (even at low concentrations), has a better effect on inducing cancer cell death in comparison to DOX alone. The result of this study suggests that the combination therapy of MB-PDT-DOX can be used as a potential strategy for the treatment of triple-negative breast cancer cells.
Collapse
Affiliation(s)
- Arghavan Yousefi Sadeghloo
- Department of Biochemistry, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran. z.kianmehr@.ut.ac.ir
| | - Khatereh Khorsandi
- Department of Biochemistry, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran. z.kianmehr@.ut.ac.ir and Department of Photodynamic, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran.
| | - Zahra Kianmehr
- Department of Biochemistry, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran. z.kianmehr@.ut.ac.ir
| |
Collapse
|
21
|
Jung E, Shim I, An J, Ji MS, Jangili P, Chi SG, Kim JS. Phenylthiourea-Conjugated BODIPY as an Efficient Photosensitizer for Tyrosinase-Positive Melanoma-Targeted Photodynamic Therapy. ACS APPLIED BIO MATERIALS 2021; 4:2120-2127. [PMID: 35014340 DOI: 10.1021/acsabm.0c01322] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Melanoma is the most threatening form of metastatic skin cancer that develops from melanocytes and causes a large majority of deaths due to poor therapeutic prognosis. It has significant limitations in treatment because it shows great resistance to chemotherapy, radiotherapy, and other therapeutic methods. A noninvasive and clinically accepted therapeutic modality, photodynamic therapy (PDT), is a promising treatment option, but it is limitedly applied for melanoma skin cancer treatment. This is because most of the photosensitizers are unlikely to be expected to have a remarkable effect on melanoma due to drug efflux by melanin pigmentation and intrinsic antioxidant defense mechanisms. Moreover, melanin is a dominant absorber in the spectral region of 500-600 nm that can cause the decreased photoreaction efficiency of photosensitizers. Herein, to overcome these drawbacks, we have developed a phenylthiourea-conjugated BODIPY photosensitizer (PTUBDP) for tyrosinase-positive melanoma-targeted PDT. In light of our results, it exhibited an enhanced cytotoxic efficacy compared to BDP, a parallel PDT agent that absence of phenylthiourea unit. PTUBDP shows outstanding effects of increased oxidative stress by an enhanced cellular uptake of the tyrosinase positive melanoma cell line (B16F10). This work presents increased therapeutic efficacy through the combined therapeutic approach, enabling enhanced reactive oxygen species (ROS) generation as well as overcoming the critical limitations of melanoma.
Collapse
Affiliation(s)
- Eugeine Jung
- Department of Chemistry, Korea University, Seoul 02841, Korea
- Department of Life Sciences, Korea University, Seoul 02841, Korea
| | - Inseob Shim
- Department of Chemistry, Korea University, Seoul 02841, Korea
| | - Jusung An
- Department of Chemistry, Korea University, Seoul 02841, Korea
| | - Myung Sun Ji
- Department of Chemistry, Korea University, Seoul 02841, Korea
| | | | - Sung-Gil Chi
- Department of Life Sciences, Korea University, Seoul 02841, Korea
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, Korea
| |
Collapse
|
22
|
Park J, Lee YK, Park IK, Hwang SR. Current Limitations and Recent Progress in Nanomedicine for Clinically Available Photodynamic Therapy. Biomedicines 2021; 9:85. [PMID: 33467201 PMCID: PMC7830249 DOI: 10.3390/biomedicines9010085] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 02/07/2023] Open
Abstract
Photodynamic therapy (PDT) using oxygen, light, and photosensitizers has been receiving great attention, because it has potential for making up for the weakness of the existing therapies such as surgery, radiation therapy, and chemotherapy. It has been mainly used to treat cancer, and clinical tests for second-generation photosensitizers with improved physicochemical properties, pharmacokinetic profiles, or singlet oxygen quantum yield have been conducted. Progress is also being made in cancer theranostics by using fluorescent signals generated by photosensitizers. In order to obtain the effective cytotoxic effects on the target cells and prevent off-target side effects, photosensitizers need to be localized to the target tissue. The use of nanocarriers combined with photosensitizers can enhance accumulation of photosensitizers in the tumor site, owing to preferential extravasation of nanoparticles into the tumor vasculature by the enhanced permeability and retention effect. Self-assembly of amphiphilic polymers provide good loading efficiency and sustained release of hydrophobic photosensitizers. In addition, prodrug nanomedicines for PDT can be activated by stimuli in the tumor site. In this review, we introduce current limitations and recent progress in nanomedicine for PDT and discuss the expected future direction of research.
Collapse
Affiliation(s)
- Jooho Park
- Department of Biomedical Chemistry, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Korea;
| | - Yong-Kyu Lee
- Department of Chemical and Biological Engineering, Korea National University of Transportation, Chungju 27469, Korea;
| | - In-Kyu Park
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun 58128, Korea;
| | - Seung Rim Hwang
- College of Pharmacy, Chosun University, Gwangju 61452, Korea
| |
Collapse
|
23
|
Effect of photodynamic therapy on expression of HRAS, NRAS and caspase 3 genes at mRNA levels, apoptosis of head and neck squamous cell carcinoma cell line. Photodiagnosis Photodyn Ther 2020; 33:102142. [PMID: 33307231 DOI: 10.1016/j.pdpdt.2020.102142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 11/14/2020] [Accepted: 11/30/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVES This study aimed to assess the effect of photodynamic therapy (PDT) on expression of CASP3, NRAS and HRAS genes at mRNA levels, and apoptosis of head and neck squamous cell carcinoma (HNSCC) cell line. MATERIALS AND METHODS In order to complete the present in vitro study, HNSCC cell line (NCBI C196 HN5) purchased from Pasteur Institute. Cells were divided into four groups; Group 1: photodynamic treatment (laser + methylene blue (MB) as photosensitizer), group 2: MB, group 3: laser (with 660 nm wavelength), and group 4: control (without any treatment). To determine the optimal concentration of MB, in a pilot study, toxicity of MB in different concentration was assessed using MTT assay. Cells in group 1, 2 and 3 was treated at optimal concentration of MB (1.6 μg/mL). Gene expression at mRNA levels was assessed after 24 h incubation, using real-time (qRT)-PCR. The expression of BAX and BCL2 genes at the mRNA levels was analyzed to evaluate apoptosis. 2-ΔΔCt values of BCL2, BAX, CASP3, NRAS, and HRAS in groups was analyzed using ANOVA. Tukey's HSD and Games Howell test was used to compare between two groups. RESULTS Over-expression of BAX (p < 0.001), CASP3 (p < 0.001) and down-regulation of BCL2 (p = 0.004), HRAS (p = 0.023) and NRAS (p = 0.045) were noted in group 1 (PDT), compared with the control group. Treatment by laser alone induce down-regulation of CASP3 (p < 0.05), BAX (p < 0.05), BCL2 (p > 0.05), HRAS (p > 0.05) and NRAS (p > 0.05). CONCLUSION PDT caused down-regulation of NRAS, HRAS and BCL2 and over-expression of CASP3 and BAX genes at mRNA levels in HNSCC cell line. The present study raises the possibility that the role of MB on BCL2 down-regulation and BAX and CASP3 over-expression was higher than laser alone while it seems that laser alone was more effective than MB in HRAS and NRAS down-regulation.
Collapse
|
24
|
Liew HS, Mai CW, Zulkefeli M, Madheswaran T, Kiew LV, Delsuc N, Low ML. Recent Emergence of Rhenium(I) Tricarbonyl Complexes as Photosensitisers for Cancer Therapy. Molecules 2020; 25:4176. [PMID: 32932573 PMCID: PMC7571230 DOI: 10.3390/molecules25184176] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 02/05/2023] Open
Abstract
Photodynamic therapy (PDT) is emerging as a significant complementary or alternative approach for cancer treatment. PDT drugs act as photosensitisers, which upon using appropriate wavelength light and in the presence of molecular oxygen, can lead to cell death. Herein, we reviewed the general characteristics of the different generation of photosensitisers. We also outlined the emergence of rhenium (Re) and more specifically, Re(I) tricarbonyl complexes as a new generation of metal-based photosensitisers for photodynamic therapy that are of great interest in multidisciplinary research. The photophysical properties and structures of Re(I) complexes discussed in this review are summarised to determine basic features and similarities among the structures that are important for their phototoxic activity and future investigations. We further examined the in vitro and in vivo efficacies of the Re(I) complexes that have been synthesised for anticancer purposes. We also discussed Re(I) complexes in conjunction with the advancement of two-photon PDT, drug combination study, nanomedicine, and photothermal therapy to overcome the limitation of such complexes, which generally absorb short wavelengths.
Collapse
Affiliation(s)
- Hui Shan Liew
- School of Postgraduate Studies, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
| | - Chun-Wai Mai
- Centre for Cancer and Stem Cell Research, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (M.Z.); (T.M.)
| | - Mohd Zulkefeli
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (M.Z.); (T.M.)
| | - Thiagarajan Madheswaran
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (M.Z.); (T.M.)
| | - Lik Voon Kiew
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Nicolas Delsuc
- Laboratoire des Biomolécules, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, 75005 Paris, France;
| | - May Lee Low
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (M.Z.); (T.M.)
| |
Collapse
|
25
|
Tang Q, Hu P, Peng H, Zhang N, Zheng Q, He Y. Near-Infrared Laser-Triggered, Self-Immolative Smart Polymersomes for in vivo Cancer Therapy. Int J Nanomedicine 2020; 15:137-149. [PMID: 32021170 PMCID: PMC6964533 DOI: 10.2147/ijn.s224502] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/14/2019] [Indexed: 12/24/2022] Open
Abstract
Purpose Traditional chemotherapy is accompanied by significant side effects, which, in many aspects, limits its treatment efficacy and clinical applications. Herein, we report an oxidative responsive polymersome nanosystem mediated by near infrared (NIR) light which exhibited the combination effect of photodynamic therapy (PDT) and chemotherapy. Methods In our study, poly (propylene sulfide)20-bl-poly (ethylene glycol)12 (PPS20-b-PEG12) block copolymer was synthesized and employed to prepare the polymersome. The hydrophobic photosensitizer zinc phthalocyanine (ZnPc) was loaded in the shell and the hydrophilic doxorubicin hydrochloride (DOX·HCl) in the inner aqueous space of the polymersome. Results Under the irradiation of 660 nm NIR light, singlet oxygen 1O2 molecules were generated from ZnPc to oxidize the neighbouring sulfur atoms on the PPS block which eventually ruptured the intact structure of polymersomes, leading to the release of encapsulated DOX·HCl. The released DOX and the 1O2 could achieve a combination effect for cancer therapy if the laser activation and drug release occur at the tumoral sites. In vitro studies confirmed the generation of singlet oxygen and DOX release by NIR irradiation. In vivo studies showed that such a combined PDT-chemotherapy nanosystem could accumulate in A375 tumors efficiently, thus leading to significant inhibition on tumor growth as compared to PDT (PZ group) or chemotherapy alone (DOX group). Conclusion In summary, this oxidation-sensitive nanosystem showed excellent anti-tumor effects by synergistic chemophotodynamic therapy, indicating that this novel drug delivery strategy could potentially provide a new means for cancer treatments in clinic.
Collapse
Affiliation(s)
- Qing Tang
- School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, People's Republic of China
| | - Ping Hu
- School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, People's Republic of China.,School of Pharmacy, Jinan University, Guangzhou 510632, People's Republic of China
| | - Haibo Peng
- School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, People's Republic of China
| | - Ning Zhang
- School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, People's Republic of China
| | - Qiang Zheng
- School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, People's Republic of China
| | - Yun He
- School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, People's Republic of China
| |
Collapse
|