1
|
Li L, Bian L, Kou N, Yuan Y, Zou H. Special immune-related adverse events and subsequent photodynamic therapy in tislelizumab treatment for esophageal cancer: a case report. Front Immunol 2024; 15:1497259. [PMID: 39654898 PMCID: PMC11625816 DOI: 10.3389/fimmu.2024.1497259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 11/07/2024] [Indexed: 12/12/2024] Open
Abstract
This case report highlights the immune-related adverse events (irAEs) that occurred during the treatment of esophageal cancer with Tislelizumab and discusses management strategies, indicating that photodynamic therapy (PDT) may be an optimal adjunctive treatment option. Following Tislelizumab therapy, the patient demonstrated significant tumor reduction; however, subsequent irAEs related to immunotherapy emerged, including eyelid muscle weakness and myocardial and skeletal muscle injury. Methylprednisolone successfully alleviated these symptoms, with early intervention being crucial for controlling irAEs. The patient then underwent PDT, which not only further helped manage irAEs but also inhibited tumor progression. This case underscores the specific adverse reactions, such as eyelid ptosis, skeletal muscle, and myocardial damage associated with Tislelizumab, and the importance of early corticosteroid intervention. It also emphasizes the significance of PDT as an adjunctive treatment for controlling tumors and alleviating immune-related adverse reactions.
Collapse
Affiliation(s)
- Longzhao Li
- Respiratory Disease Center, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Integrative Traditional Chinese and Western Medicine, Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Lingjie Bian
- Respiratory Disease Center, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Na Kou
- Respiratory Disease Center, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yue Yuan
- Respiratory Disease Center, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Heng Zou
- Respiratory Disease Center, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
2
|
Giram P, Bist G, Woo S, Wohlfert E, Pili R, You Y. Prodrugs of paclitaxel improve in situ photo-vaccination. Photochem Photobiol 2024:10.1111/php.14025. [PMID: 39384406 PMCID: PMC11978925 DOI: 10.1111/php.14025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/25/2024] [Accepted: 08/22/2024] [Indexed: 10/11/2024]
Abstract
Photodynamic therapy (PDT) effectively kills cancer cells and initiates immune responses that promote anticancer effects locally and systemically. Primarily developed for local and regional cancers, the potential of PDT for systemic antitumor effects [in situ photo-vaccination (ISPV)] remains underexplored. This study investigates: (1) the comparative effectiveness of paclitaxel (PTX) prodrug [Pc-(L-PTX)2] for PDT and site-specific PTX effects versus its pseudo-prodrug [Pc-(NCL-PTX)2] for PDT combined with checkpoint inhibitors; (2) mechanisms driving systemic antitumor effects; and (3) the prophylactic impact on preventing cancer recurrence. A bilateral tumor model was established in BALB/c mice through subcutaneous injection of CT26 cells. Mice received the PTX prodrug (0.5 μmole kg-1, i.v.), and tumors were treated with a 690-nm laser (75 mW cm-2 for 30 min, drug-light interval 0.5 h, light does 135 J cm-1), followed by anti-CTLA-4 (100 μg dose-1, i.p.) on days 1, 4, and 7. Notable enhancement in both local and systemic antitumor effectiveness was observed with [Pc-(L-PTX)2] compared to [Pc-(NCL-PTX)2] with checkpoint inhibitor. Immune cell depletion and immunohistochemistry confirmed neutrophils and CD8+ T cells are effectors for systemic antitumor effects. Treatment-induced immune memory resisted newly rechallenged CT26, showcasing prophylactic benefits. ISPV with a PTX prodrug and anti-CTLA-4 is a promising approach for treating metastatic cancers and preventing recurrence.
Collapse
Affiliation(s)
- Prabhanjan Giram
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Ganesh Bist
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Sukyung Woo
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Elizabeth Wohlfert
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY,14203, USA
| | - Roberto Pili
- Division of Hematology and Oncology, Department of Medicine, University at Buffalo, Buffalo, NY, 14203, USA
| | - Youngjae You
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| |
Collapse
|
3
|
Han J, Xu X, Jin F, Xu X, Fang T, Du Y. Tumor oxygenation nanoliposomes promote deep photodynamic therapy for triple-negative breast cancer. Biomater Sci 2024; 12:4967-4979. [PMID: 39158634 DOI: 10.1039/d4bm00847b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive type of breast cancer and has many characteristics including high metastatic rates, poor overall survival, and low response to traditional chemotherapy. Photodynamic therapy (PDT), emerging as a precise treatment modality, has shown promise in improving the antitumor response. However, it still faces challenges such as limited light penetration depth, rapid oxygen consumption, and inadequate targeting ability. In this study, we developed Rose Bengal (RB, photosensitizer) and oxygen co-loaded CREKA-modified UCNP-based nanoliposomes (CLIP-RB-PFOB@UCNP) for tumor targeting and near-infrared (NIR)-triggered deep and long-lasting PDT. Our results demonstrated that CLIP-RB-PFOB@UCNP effectively targeted and accumulated in tumor tissue through the interaction between CREKA and fibronectin, which is overexpressed in tumor cells. Under NIR irradiation, CLIP-RB-PFOB@UCNP exhibited significant destruction of orthotopic tumors, reduced the level of HIF-1α, and efficiently suppressed lung metastasis in a metastatic TNBC model. In conclusion, this study offers new avenues for improving the therapeutic outcomes of PDT for clinical TNBC treatment.
Collapse
Affiliation(s)
- Jianhua Han
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China.
| | - Xinyi Xu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China.
| | - Feiyang Jin
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China.
| | - Xiaoling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang 310015, P. R. China.
| | - Tao Fang
- Department of Anesthesiology, The Affiliated Jinhua Hospital of Wenzhou Medical University, Jinhua, Zhejiang 321000, P. R. China.
| | - Yongzhong Du
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China.
- Innovation Center of Translational Pharmacy, Jinhua Institute of Zhejiang University, Jinhua 321299, P. R. China
| |
Collapse
|
4
|
Li Y, Li Y, Song Y, Liu S. Advances in research and application of photodynamic therapy in cholangiocarcinoma (Review). Oncol Rep 2024; 51:53. [PMID: 38334150 DOI: 10.3892/or.2024.8712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/18/2024] [Indexed: 02/10/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a disease characterized by insidious clinical manifestations and challenging to diagnose. Patients are usually diagnosed at an advanced stage and miss the opportunity for radical surgery. Therefore, effective palliative therapy is the main treatment approach for unresectable CCA. Current common palliative treatments include biliary drainage, chemotherapy, radiotherapy, targeted therapy and immunotherapy. However, these treatments only offer limited improvement in quality of life and survival. Photodynamic therapy (PDT) is a novel local treatment method that is considered a safe tumor ablation method for numerous cancers. It has shown good efficacy in various studies of CCA and is expected to become an important treatment for CCA. In the present study, the mechanisms of PDT in the treatment of CCA were systematically explored and the progress in the research of photosensitizers was discussed. The current study focused on the various PDT protocols and their therapeutic effects in CCA, with the objective of providing a new horizon for future research and clinical applications of PDT in the treatment of CCA.
Collapse
Affiliation(s)
- Yufeng Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, P.R. China
| | - Yuhang Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, P.R. China
| | - Yinghui Song
- Central Laboratory of Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan 410005, P.R. China
| | - Sulai Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, P.R. China
| |
Collapse
|
5
|
Gao M, Guo H, Dong X, Wang Z, Yang Z, Shang Q, Wang Q. Regulation of inflammation during wound healing: the function of mesenchymal stem cells and strategies for therapeutic enhancement. Front Pharmacol 2024; 15:1345779. [PMID: 38425646 PMCID: PMC10901993 DOI: 10.3389/fphar.2024.1345779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/05/2024] [Indexed: 03/02/2024] Open
Abstract
A wound takes a long time to heal and involves several steps. Following tissue injury, inflammation is the primary cause of tissue regeneration and repair processes. As a result, the pathophysiological processes involving skin damage, healing, and remodeling depend critically on the control of inflammation. The fact that it is a feasible target for improving the prognosis of wound healing has lately become clear. Mesenchymal stem cells (MSCs) are an innovative and effective therapeutic option for wound healing due to their immunomodulatory and paracrine properties. By controlling the inflammatory milieu of wounds through immunomodulation, transplanted MSCs have been shown to speed up the healing process. In addition to other immunomodulatory mechanisms, including handling neutrophil activity and modifying macrophage polarization, there may be modifications to the activation of T cells, natural killer (NK) cells, and dendritic cells (DCs). Furthermore, several studies have shown that pretreating MSCs improves their ability to modulate immunity. In this review, we summarize the existing knowledge about how MSCs influence local inflammation in wounds by influencing immunity to facilitate the healing process. We also provide an overview of MSCs optimizing techniques when used to treat wounds.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qiying Wang
- Department of Plastic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
6
|
Baydoun M, Boidin L, Leroux B, Vignion-Dewalle AS, Quilbe A, Grolez GP, Azaïs H, Frochot C, Moralès O, Delhem N. Folate Receptor Targeted Photodynamic Therapy: A Novel Way to Stimulate Anti-Tumor Immune Response in Intraperitoneal Ovarian Cancer. Int J Mol Sci 2023; 24:11288. [PMID: 37511049 PMCID: PMC10378870 DOI: 10.3390/ijms241411288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/20/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Photodynamic therapy (PDT) has shown improvements in cancer treatment and in the induction of a proper anti-tumor immune response. However, current photosensitizers (PS) lack tumor specificity, resulting in reduced efficacy and side effects in patients with intraperitoneal ovarian cancer (OC). In order to target peritoneal metastases of OC, which overexpress folate receptor (FRα) in 80% of cases, we proposed a targeted PDT using a PS coupled with folic acid. Herein, we applied this targeted PDT in an in vivo mouse model of peritoneal ovarian carcinomatosis. The efficacy of the treatment was evaluated in mice without and with human peripheral blood mononuclear cell (PBMC) reconstitution. When mice were reconstituted, using a fractionized PDT protocol led to a significantly higher decrease in the tumor growth than that obtained in the non-reconstituted mice (p = 0.0469). Simultaneously, an immune response was reflected by an increase in NK cells, and both CD4+ and CD8+ T cells were activated. A promotion in cytokines IFNγ and TNFα and an inhibition in cytokines TGFβ, IL-8, and IL-10 was also noticed. Our work showed that a fractionized FRα-targeted PDT protocol is effective for the treatment of OC and goes beyond local induction of tumor cell death, with the promotion of a subsequent anti-tumor response.
Collapse
Affiliation(s)
- Martha Baydoun
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, 59000 Lille, France
| | - Léa Boidin
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, 59000 Lille, France
| | - Bertrand Leroux
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, 59000 Lille, France
| | - Anne-Sophie Vignion-Dewalle
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, 59000 Lille, France
| | - Alexandre Quilbe
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, 59000 Lille, France
| | - Guillaume Paul Grolez
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, 59000 Lille, France
| | - Henri Azaïs
- Department of Gynecological and Breast Surgery and Oncology, Assistance Publique-Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière University Hospital, 75013 Paris, France
| | - Céline Frochot
- Laboratoire des Réactions et Génie des Procédés (LRGP), CNRS-Université de Lorraine, 1 Rue Grandville, 54000 Nancy, France
| | - Olivier Moralès
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, 59000 Lille, France
- INSERM UMR9020-UMR-S 1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France
| | - Nadira Delhem
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, 59000 Lille, France
| |
Collapse
|
7
|
Olek M, Machorowska-Pieniążek A, Czuba ZP, Cieślar G, Kawczyk-Krupka A. Effect of Hypericin-Mediated Photodynamic Therapy on the Secretion of Soluble TNF Receptors by Oral Cancer Cells. Pharmaceutics 2023; 15:pharmaceutics15041279. [PMID: 37111765 PMCID: PMC10140873 DOI: 10.3390/pharmaceutics15041279] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Squamous cell carcinoma is the most common cancer of the head and neck region. In addition to the classic surgical treatment method, alternative therapy methods are sought. One such method is photodynamic therapy (PDT). In addition to the direct cytotoxic effect, it is essential to determine the effect of PDT on persistent tumor cells. The study used the SCC-25 oral squamous cell carcinoma (OSCC) cell line and the HGF-1 healthy gingival fibroblast line. A compound of natural origin-hypericin (HY)-was used as a photosensitizer (PS) at concentrations of 0-1 µM. After two hours of incubation with the PS, the cells were irradiated with light doses of 0-20 J/cm2. The 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) test was used to determine sublethal doses of PDT. Cell supernatants subjected to sublethal PDT were assessed for soluble tumor necrosis alpha receptors (sTNF-R1, sTNF-R2). The phototoxic effect was observed starting with a light dose of 5 J/cm2 and amplified with the increase in HY concentration and light dose. A statistically significant increase in sTNF-R1 secretion by SCC-25 cells was demonstrated after the PDT with 0.5 µM HY and irradiation with 2 J/cm2 (sTNF-R1 concentration = 189.19 pg/mL ± 2.60) compared to the control without HY and irradiated with the same dose of light (sTNF-R1 concentration = 108.94 pg/mL ± 0.99). The baseline production of sTNF-R1 was lower for HGF-1 than for SCC-25, and secretion was not affected by the PDT. The PDT had no effect on the sTNF-R2 production in the SCC-25 or HGF-1 lines.
Collapse
Affiliation(s)
- Marcin Olek
- Doctoral School of Medical University of Silesia, 40-055 Katowice, Poland
- Department of Orthodontics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland
| | | | - Zenon P Czuba
- Department of Microbiology and Immunology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland
| | - Grzegorz Cieślar
- Department of Internal Diseases, Angiology and Physical Medicine, Center for Laser Diagnostics and Therapy, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland
| | - Aleksandra Kawczyk-Krupka
- Department of Internal Diseases, Angiology and Physical Medicine, Center for Laser Diagnostics and Therapy, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland
| |
Collapse
|
8
|
He L, Xu F, Li Y, Jin H, Lo PC. Cupric-ion-promoted fabrication of oxygen-replenishing nanotherapeutics for synergistic chemo and photodynamic therapy against tumor hypoxia. Acta Biomater 2023; 162:57-71. [PMID: 36944404 DOI: 10.1016/j.actbio.2023.03.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/16/2023] [Accepted: 03/13/2023] [Indexed: 03/23/2023]
Abstract
Mixing a glutathione (GSH)-responsive carboxy zinc(II) phthalocyanine (ZnPc*) and CuSO4·5H2O in water with or without the presence of the anticancer drug SN38 resulted in the formation of self-assembled nanotherapeutics labeled as ZnPc*/Cu/SN38@NP and ZnPc*/Cu@NP, respectively. The Cu2+ ions not only promoted the self-assembly of the carboxy phthalocyanine through metal complexation, but also catalyzed the transformation of H2O2 to oxygen via a catalase-like reaction, rendering an oxygen-replenishing property to the nanosystems. Both nanosystems exhibited high stability in aqueous media, but the nanoparticles disassembled gradually in an acidic or GSH-enriched environment and inside human colorectal adenocarcinoma HT29 cells, releasing the encapsulated therapeutic components. The disassembly process together with the activation by the intracellular GSH led to relaxation of the intrinsic quenching of the nanophotosensitizers and restoration of the photoactivities of ZnPc*. Under a hypoxic condition, ZnPc*/Cu/SN38@NP could attenuate the intracellular hypoxia level and maintain the photodynamic activity due to its Cu2+-promoted oxygen-replenishing ability. The photodynamic effect of ZnPc* and the anticancer effect of SN38 worked cooperatively, causing substantial apoptotic cell death. The dual therapeutic actions could also effectively inhibit the tumor growth in HT29 tumor-bearing nude mice without initiating notable adverse effects to the mice. STATEMENT OF SIGNIFICANCE: The oxygen-dependent nature of photodynamic therapy generally reduces its efficacy against tumor hypoxia, which is a common characteristic of advanced solid tumors and usually leads to resistance toward various anticancer therapies. We report herein a facile approach to assemble a glutathione-responsive carboxy phthalocyanine-based photosensitizer and an anticancer drug in aqueous media, in which Cu(II) ions were used to promote the self-assembly through metal complexation and catalyze the conversion of H2O2 to oxygen through a catalase-like reaction, making the resulting nanoparticles possessing an oxygen-replenishing property that could promote the photodynamic effect against hypoxic cancer cells and tumors. The use of Cu(II) ions to achieve the aforementioned dual functions in the fabrication of advanced nano-photosensitizing systems has not been reported.
Collapse
Affiliation(s)
- Lin He
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Feijie Xu
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Yongxin Li
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Honglin Jin
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Pui-Chi Lo
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China.
| |
Collapse
|
9
|
Girotti AW, Fahey JF, Korytowski W. Role of nitric oxide in hyper-aggressiveness of tumor cells that survive various anti-cancer therapies. Crit Rev Oncol Hematol 2022; 179:103805. [PMID: 36087851 DOI: 10.1016/j.critrevonc.2022.103805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/10/2022] [Accepted: 09/05/2022] [Indexed: 11/18/2022] Open
Abstract
Low level nitric oxide (NO) produced by inducible NO synthase (iNOS) in many malignant tumors is known to play a key role in the survival and proliferation of tumor cells. NO can also induce or augment resistance to anti-tumor treatments such as platinum-based chemotherapy (CT), ionizing radiotherapy (RT), and non-ionizing photodynamic therapy (PDT). In each of these treatments, tumor cells that survive the challenge may exhibit a striking increase in NO-dependent proliferative, migratory, and invasive aggressiveness compared with non-challenged controls. Moreover, NO from cells directly targeted by PDT can often stimulate aggressiveness in non- or poorly targeted bystander cells. Although NO-mediated resistance to many of these therapies is fairly-well recognized by now, the hyper-aggressiveness of surviving cells and bystander counterparts is not. We will focus on these negative aspects in this review, citing examples from the PDT, CT, and RT publications. Increased aggressiveness of cells that escape therapeutic elimination is a concern because it could enhance tumor progression and metastatic dissemination. Pharmacologic approaches for suppressing these negative responses will also be discussed, e.g., administering inhibitors of iNOS activity or iNOS expression as therapeutic adjuvants.
Collapse
Affiliation(s)
- Albert W Girotti
- Depatrment of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Jonathan F Fahey
- Department of Pathology, University of Colorado, Aurora, CO, USA
| | | |
Collapse
|
10
|
Potential of Black Phosphorus in Immune-Based Therapeutic Strategies. Bioinorg Chem Appl 2022; 2022:3790097. [PMID: 35859703 PMCID: PMC9293569 DOI: 10.1155/2022/3790097] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/07/2022] [Accepted: 03/31/2022] [Indexed: 12/03/2022] Open
Abstract
Black phosphorus (BP) consists of phosphorus atoms, an essential element of bone and nucleic acid, which covalently bonds to three adjacent phosphorus atoms to form a puckered bilayer structure. With its anisotropy, band gap, biodegradability, and biocompatibility properties, BP is considered promising for cancer therapy. For example, BP under irradiation can convert near-infrared (NIR) light into heat and reactive oxygen species (ROS) to damage cancer cells, called photothermal therapy (PTT) and photodynamic therapy (PDT). Compared with PTT and PDT, the novel techniques of sonodynamic therapy (SDT) and photoacoustic therapy (PAT) exhibit amplified ROS generation and precise photoacoustic-shockwaves to enhance anticancer effect when BP receives ultrasound or NIR irradiation. Based on the prospective phototherapy, BP with irradiation can cause a “double-kill” to tumor cells, involving tumor-structure damage induced by heat, ROS, and shockwaves and a subsequent anticancer immune response induced by in situ vaccines construction in tumor site, which is referred to as photoimmunotherapy (PIT). In conclusion, BP shows promise in natural antitumor biological activity, biological imaging, drug delivery, PTT/PDT/SDT/PAT/PIT, nanovaccines, nanoadjuvants, and combination immunotherapy regimens.
Collapse
|
11
|
Yan J, Lai H, Zeng B, Wang L, Xing F, Xiao P. Photoinduced free radical-releasing systems and their anticancer properties. Photochem Photobiol Sci 2022; 21:1405-1417. [PMID: 35553411 DOI: 10.1007/s43630-022-00231-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 04/11/2022] [Indexed: 11/26/2022]
Abstract
Cancer has been a serious threat and impact on the health and life of human. Phototherapy is considered as a promising therapeutic method to replace the traditional treatment in clinic owing to its noninvasive nature and high efficiency. Photoinitiators have long been used in the field of photopolymerization; however, few studies have been carried out on their potential as anticancer agents under light irradiation. In this study, the effect of a photoinitiator, diphenyl (2, 4, 6-trimethylbenzoyl) phosphine oxide (TPO), on breast cancer is investigated and the related mechanism is elucidated. It is found that TPO has low dark toxicity and significant phototoxicity. TPO can inhibit cell growth and development and promote cell apoptosis through a mitochondrial pathway under light irradiation. Further studies show that cell apoptosis is induced by free radicals produced from the photolysis of TPO to activate JNK phosphorylation. Overall, we identify the antitumor effects of TPO in vitro for the first time, and provides a proof of concept for its application as a novel photolatent therapeutic drug.
Collapse
Affiliation(s)
- Jieyu Yan
- Department of Immunobiology, College of Life Science and Technology, Jinan University, #601 Huangpu West Avenue, Guangzhou, 510632, China
| | - Haiwang Lai
- Department of Immunobiology, College of Life Science and Technology, Jinan University, #601 Huangpu West Avenue, Guangzhou, 510632, China.
| | - Boning Zeng
- Department of Immunobiology, College of Life Science and Technology, Jinan University, #601 Huangpu West Avenue, Guangzhou, 510632, China
| | - Liqing Wang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, #601 Huangpu West Avenue, Guangzhou, 510632, China
| | - Feiyue Xing
- Department of Immunobiology, College of Life Science and Technology, Jinan University, #601 Huangpu West Avenue, Guangzhou, 510632, China.
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, China.
| | - Pu Xiao
- Research School of Chemistry, The Australian National University, Canberra, ACT, 2601, Australia.
| |
Collapse
|
12
|
Ma Y, Xiao F, Lu C, Wen L. Multifunctional Nanosystems Powered Photodynamic Immunotherapy. Front Pharmacol 2022; 13:905078. [PMID: 35645842 PMCID: PMC9130658 DOI: 10.3389/fphar.2022.905078] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 04/25/2022] [Indexed: 12/19/2022] Open
Abstract
Photodynamic Therapy (PDT) with the intrinsic advantages including non-invasiveness, spatiotemporal selectivity, low side-effects, and immune activation ability has been clinically approved for the treatment of head and neck cancer, esophageal cancer, pancreatic cancer, prostate cancer, and esophageal squamous cell carcinoma. Nevertheless, the PDT is only a strategy for local control of primary tumor, that it is hard to remove the residual tumor cells and inhibit the tumor metastasis. Recently, various smart nanomedicine-based strategies are developed to overcome the barriers of traditional PDT including the drawbacks of traditional photosensitizers, limited tissue penetrability of light, inefficient induction of tumor cell death and tumor resistance to the therapy. More notably, a growing number of studies have focused on improving the therapeutic efficiency by eliciting host immune system with versatile nanoplatforms, which heralds a broader clinical application prospect of PDT in the future. Herein, the pathways of PDT induced-tumor destruction, especially the host immune response is summarized, and focusing on the recent progress of nanosystems-enhanced PDT through eliciting innate immunity and adaptive immunity. We expect it will provide some insights for conquering the drawbacks current PDT and expand the range of clinical application through this review.
Collapse
Affiliation(s)
- Yunong Ma
- Medical College, Guangxi University, Nanning, China
- Zhuhai Precision Medical Center, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated With Jinan University), Jinan University, Zhuhai, China
| | - Fengfeng Xiao
- Zhuhai Precision Medical Center, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated With Jinan University), Jinan University, Zhuhai, China
| | - Cuixia Lu
- Medical College, Guangxi University, Nanning, China
- *Correspondence: Cuixia Lu, ; Liewei Wen,
| | - Liewei Wen
- Zhuhai Precision Medical Center, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated With Jinan University), Jinan University, Zhuhai, China
- *Correspondence: Cuixia Lu, ; Liewei Wen,
| |
Collapse
|
13
|
Lin S, Liu C, Zhao X, Han X, Li X, Ye Y, Li Z. Recent Advances of Pyridinone in Medicinal Chemistry. Front Chem 2022; 10:869860. [PMID: 35402370 PMCID: PMC8984125 DOI: 10.3389/fchem.2022.869860] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 03/02/2022] [Indexed: 12/11/2022] Open
Abstract
Pyridinones have been adopted as an important block in medicinal chemistry that could serve as hydrogen bond donors and acceptors. With the help of feasible synthesis routes via established condensation reactions, the physicochemical properties of such a scaffold could be manipulated by adjustment of polarity, lipophilicity, and hydrogen bonding, and eventually lead to its wide application in fragment-based drug design, biomolecular mimetics, and kinase hinge-binding motifs. In addition, most pyridinone derivatives exhibit various biological activities ranging from antitumor, antimicrobial, anti-inflammatory, and anticoagulant to cardiotonic effects. This review focuses on recent contributions of pyridinone cores to medicinal chemistry, and addresses the structural features and structure–activity relationships (SARs) of each drug-like molecule. These advancements contribute to an in-depth understanding of the potential of this biologically enriched scaffold and expedite the development of its new applications in drug discovery.
Collapse
Affiliation(s)
- Shibo Lin
- Department of Pharmacy, Chengdu Second People’s Hospital, Chengdu, China
- *Correspondence: Shibo Lin,
| | - Chun Liu
- Department of Pharmacy, Chengdu Second People’s Hospital, Chengdu, China
| | - Xiaotian Zhao
- Department of Pharmacy, Chengdu Second People’s Hospital, Chengdu, China
| | - Xiao Han
- Department of Pharmacy, Chengdu Second People’s Hospital, Chengdu, China
| | - Xuanhao Li
- Department of Pharmacy, Chengdu Second People’s Hospital, Chengdu, China
| | - Yongqin Ye
- Department of Pharmacy, Chengdu Second People’s Hospital, Chengdu, China
| | - Zheyu Li
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, China
| |
Collapse
|
14
|
Lipid-based nanoparticles for photosensitive drug delivery systems. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2022; 52:151-160. [PMID: 35013696 PMCID: PMC8731178 DOI: 10.1007/s40005-021-00553-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/28/2021] [Indexed: 12/16/2022]
Abstract
Background Numerous drug delivery strategies have been studied, but many hurdles exist in drug delivery rates to the target site. Recently, researchers have attempted to remotely control the in vivo behavior of drugs with light to overcome the shortcomings of conventional drug delivery systems. Photodynamic and photothermal systems are representative strategies wherein a photosensitive material is activated in response to a specific wavelength of light. Area covered Photosensitive materials generally exhibit poor solubility and low biocompatibility. Additionally, their low photostability negatively affects delivery performance. A formulation of lipid-based nanoparticles containing photosensitive substances can help achieve photosensitive drug delivery with improved biocompatibility. The lipid bilayer structure, which can be assembled and disassembled by modulating the surrounding conditions (temperature, pH, etc.), can also be crucial for controlled release of drugs. Expert opinion To the best of our knowledge, translation research on photoresponsive nanoparticles is scarce. However, as various drugs based on lipid nanoparticles have been clinically approved, the development potential of the lipid-based photoresponsive nanoparticles seems high. Thus, the identification of valid indications and development of optimum medical devices will increase the interest in photoresponsive material-based nanoparticles.
Collapse
|
15
|
Zhang D, Xie Q, Liu Y, Li Z, Li H, Li S, Li Z, Cui J, Su M, Jiang X, Xue P, Bai M. Photosensitizer IR700DX-6T- and IR700DX-mbc94-mediated photodynamic therapy markedly elicits anticancer immune responses during treatment of pancreatic cancer. Pharmacol Res 2021; 172:105811. [PMID: 34390852 DOI: 10.1016/j.phrs.2021.105811] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/26/2021] [Accepted: 08/10/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND/AIMS IR700DX-6T and IR700DX-mbc94 are two chemically synthesized photosensitizers (PSs) that target the translocator protein (TSPO) and type 2 cannabinoid receptor (CB2R), respectively, for photodynamic therapy (PDT) of cancer. Recently, we found that IR700DX-6T and IR700DX-mbc94 exhibited high selectivity and efficiency in PDT for breast cancer and malignant astrocytoma. Yet, the phototherapeutic effects of the PSs on pancreatic cancer and underlying mechanisms remain unknown. This study investigated the effect of IR700DX-6T- or IR700DX-mbc94-PDT on pancreatic cancer and whether the treatment involves eliciting anticancer immune responses in support of superior therapeutic efficacy. METHODS Four pancreatic cancer cell lines were used for in vitro studies. C57BL/6 mice bearing pancreatic cancer cell-derived xenografts were generated for in vivo studies regarding the therapeutic effects of IR700DX-6T-PDT and IR700DX-mbc94-PDT on pancreatic cancer. The immunostimulatory or immunosuppressive effects of IR700DX-6T-PDT and IR700DX-mbc94-PDT were examined by detecting CD8+ T cells, regulatory T cells (Tregs), and dendritic cells (DCs) using flow cytometry and immunohistochemistry (IHC). RESULTS TSPO and CB2R were markedly upregulated in pancreatic cancer cells and tissues. Both IR700DX-6T-PDT and IR700DX-mbc94-PDT significantly inhibited pancreatic cancer cell growth in a dose- and time-dependent manner. Notably, assessment of anticancer immune responses revealed that both IR700DX-6T-PDT and IR700DX-mbc94-PDT significantly induced CD8+ T cells, promoted maturation of DCs, and suppressed Tregs, with stronger effects exerted by IR700DX-6T-PDT compared to IR700DX-mbc94-PDT. CONCLUSIONS IR700DX-6T-PDT and IR700DX-mbc94-PDT involves eliciting anticancer immune responses. Our study has also implicated that PDT in combination with immunotherapy holds promise to improve therapeutic efficacy for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Dawei Zhang
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, China; Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Qing Xie
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Yang Liu
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Zongyan Li
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Haiyan Li
- Department of Breast Surgery, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510650, China
| | - Shiying Li
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Zhen Li
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Fourth General Surgery Department, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110000, China
| | - Jing Cui
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Nuclear Medicine, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Meng Su
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Xiaofeng Jiang
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Ping Xue
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, China.
| | - Mingfeng Bai
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
16
|
Algorri JF, Ochoa M, Roldán-Varona P, Rodríguez-Cobo L, López-Higuera JM. Photodynamic Therapy: A Compendium of Latest Reviews. Cancers (Basel) 2021; 13:4447. [PMID: 34503255 PMCID: PMC8430498 DOI: 10.3390/cancers13174447] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 12/15/2022] Open
Abstract
Photodynamic therapy (PDT) is a promising therapy against cancer. Even though it has been investigated for more than 100 years, scientific publications have grown exponentially in the last two decades. For this reason, we present a brief compendium of reviews of the last two decades classified under different topics, namely, overviews, reviews about specific cancers, and meta-analyses of photosensitisers, PDT mechanisms, dosimetry, and light sources. The key issues and main conclusions are summarized, including ways and means to improve therapy and outcomes. Due to the broad scope of this work and it being the first time that a compendium of the latest reviews has been performed for PDT, it may be of interest to a wide audience.
Collapse
Affiliation(s)
- José Francisco Algorri
- Photonics Engineering Group, University of Cantabria, 39005 Santander, Spain; (M.O.); (P.R.-V.); (J.M.L.-H.)
- CIBER-bbn, Institute of Health Carlos III, 28029 Madrid, Spain;
- Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain
| | - Mario Ochoa
- Photonics Engineering Group, University of Cantabria, 39005 Santander, Spain; (M.O.); (P.R.-V.); (J.M.L.-H.)
- CIBER-bbn, Institute of Health Carlos III, 28029 Madrid, Spain;
- Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain
| | - Pablo Roldán-Varona
- Photonics Engineering Group, University of Cantabria, 39005 Santander, Spain; (M.O.); (P.R.-V.); (J.M.L.-H.)
- CIBER-bbn, Institute of Health Carlos III, 28029 Madrid, Spain;
- Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain
| | | | - José Miguel López-Higuera
- Photonics Engineering Group, University of Cantabria, 39005 Santander, Spain; (M.O.); (P.R.-V.); (J.M.L.-H.)
- CIBER-bbn, Institute of Health Carlos III, 28029 Madrid, Spain;
- Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39011 Santander, Spain
| |
Collapse
|
17
|
Gunaydin G, Gedik ME, Ayan S. Photodynamic Therapy for the Treatment and Diagnosis of Cancer-A Review of the Current Clinical Status. Front Chem 2021; 9:686303. [PMID: 34409014 PMCID: PMC8365093 DOI: 10.3389/fchem.2021.686303] [Citation(s) in RCA: 196] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/19/2021] [Indexed: 12/24/2022] Open
Abstract
Photodynamic therapy (PDT) has been used as an anti-tumor treatment method for a long time and photosensitizers (PS) can be used in various types of tumors. Originally, light is an effective tool that has been used in the treatment of diseases for ages. The effects of combination of specific dyes with light illumination was demonstrated at the beginning of 20th century and novel PDT approaches have been developed ever since. Main strategies of current studies are to reduce off-target effects and improve pharmacokinetic properties. Given the high interest and vast literature about the topic, approval of PDT as the first drug/device combination by the FDA should come as no surprise. PDT consists of two stages of treatment, combining light energy with a PS in order to destruct tumor cells after activation by light. In general, PDT has fewer side effects and toxicity than chemotherapy and/or radiotherapy. In addition to the purpose of treatment, several types of PSs can be used for diagnostic purposes for tumors. Such approaches are called photodynamic diagnosis (PDD). In this Review, we provide a general overview of the clinical applications of PDT in cancer, including the diagnostic and therapeutic approaches. Assessment of PDT therapeutic efficacy in the clinic will be discussed, since identifying predictors to determine the response to treatment is crucial. In addition, examples of PDT in various types of tumors will be discussed. Furthermore, combination of PDT with other therapy modalities such as chemotherapy, radiotherapy, surgery and immunotherapy will be emphasized, since such approaches seem to be promising in terms of enhancing effectiveness against tumor. The combination of PDT with other treatments may yield better results than by single treatments. Moreover, the utilization of lower doses in a combination therapy setting may cause less side effects and better results than single therapy. A better understanding of the effectiveness of PDT in a combination setting in the clinic as well as the optimization of such complex multimodal treatments may expand the clinical applications of PDT.
Collapse
Affiliation(s)
- Gurcan Gunaydin
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - M. Emre Gedik
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Seylan Ayan
- Department of Chemistry, Bilkent University, Ankara, Turkey
| |
Collapse
|
18
|
Yuan G, Yao M, Lv H, Jia X, Chen J, Xue J. Novel Targeted Photosensitizer as an Immunomodulator for Highly Efficient Therapy of T-Cell Acute Lymphoblastic Leukemia. J Med Chem 2020; 63:15655-15667. [PMID: 33300796 DOI: 10.1021/acs.jmedchem.0c01072] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Dasatinib is a kinase-targeted drug used in the treatment of leukemia. Regrettably, it remains far from optimal medicine due to insurmountable drug resistance and side effects. Photodynamic therapy (PDT) has proven that it can induce systemic immune responses. However, conventional photosensitizers as immunomodulators produce anticancer immunities, which are inadequate to eliminate residual cancer cells. Herein, a novel compound 4 was synthesized and investigated, which introduces dasatinib and zinc(II) phthalocyanine as the targeting and photodynamic moiety, respectively. Compound 4 exhibits a high affinity to CCRF-CEM cells/tumor tissues, which overexpress lymphocyte-specific protein tyrosine kinase (LCK), and preferential elimination from the body. Meanwhile, compound 4 shows excellent photocytotoxicity and tumor regression. Significantly, compound 4-induced PDT can obviously enhance immune responses, resulting in the production of more immune cells. We believe that the proposed manner is a potential strategy for the treatment of T-cell acute lymphoblastic leukemia.
Collapse
Affiliation(s)
- Gankun Yuan
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 2 Xueyuan Road, University Town, Fuzhou 350116, Fujian, P. R. China
| | - Mengyu Yao
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 2 Xueyuan Road, University Town, Fuzhou 350116, Fujian, P. R. China
| | - Huihui Lv
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 2 Xueyuan Road, University Town, Fuzhou 350116, Fujian, P. R. China
| | - Xiao Jia
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 2 Xueyuan Road, University Town, Fuzhou 350116, Fujian, P. R. China
| | - Juanjuan Chen
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 2 Xueyuan Road, University Town, Fuzhou 350116, Fujian, P. R. China
| | - Jinping Xue
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 2 Xueyuan Road, University Town, Fuzhou 350116, Fujian, P. R. China
| |
Collapse
|
19
|
Yang T, Tan Y, Zhang W, Yang W, Luo J, Chen L, Liu H, Yang G, Lei X. Effects of ALA-PDT on the Healing of Mouse Skin Wounds Infected With Pseudomonas aeruginosa and Its Related Mechanisms. Front Cell Dev Biol 2020; 8:585132. [PMID: 33344449 PMCID: PMC7746815 DOI: 10.3389/fcell.2020.585132] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 11/11/2020] [Indexed: 11/24/2022] Open
Abstract
Photodynamic therapy (PDT) is a promising new method to eliminate microbial infection and promote wound healing. Its effectiveness has been confirmed by some studies; however, the mechanisms of PDT in wound healing remain obscure. We used mouse skin wounds infected with Pseudomonas aeruginosa as a research object to explore the therapeutic effects and mechanisms of 5-aminolevulinic acid photodynamic therapy (ALA-PDT). ALA-PDT treatment significantly reduced the load of P. aeruginosa in the wound and surrounding tissues and promoted the healing of skin wounds in mice. Hematoxylin-eosin (HE) and Sirius red staining showed that ALA-PDT promoted granulation tissue formation, angiogenesis, and collagen regeneration and remodeling. After ALA-PDT treatment, the expression of inflammatory factors (TNF-α and IL-1β) first increased and then decreased, while the secretion of growth factors (TGF-β-1 and VEGF) increased gradually after treatment. Furthermore, ALA-PDT affected the polarization state of macrophages, activating and promoting macrophages from an M1 to an M2 phenotype. In conclusion, ALA-PDT can not only kill bacteria but also promote wound healing by regulating inflammatory factors, collagen remodeling and macrophages. This study further clarifies the mechanism of PDT in the healing of infectious skin wounds and provides further experimental evidence for its clinical treatment of skin wounds infected by P. aeruginosa.
Collapse
Affiliation(s)
- Tao Yang
- Department of Dermatology, Daping Hospital, The Army Medical University, Chongqing, China
| | - Yang Tan
- Department of Dermatology, Daping Hospital, The Army Medical University, Chongqing, China
| | - Wentao Zhang
- Department of Dermatology, Daping Hospital, The Army Medical University, Chongqing, China
| | - Weijiang Yang
- Department of Dermatology, Daping Hospital, The Army Medical University, Chongqing, China
| | - Jiefu Luo
- Department of Dermatology, Daping Hospital, The Army Medical University, Chongqing, China
| | - Ling Chen
- Department of Dermatology, Daping Hospital, The Army Medical University, Chongqing, China
| | - Hong Liu
- Department of Dermatology, Daping Hospital, The Army Medical University, Chongqing, China
| | - Guihong Yang
- Department of Dermatology, Daping Hospital, The Army Medical University, Chongqing, China
| | - Xia Lei
- Department of Dermatology, Daping Hospital, The Army Medical University, Chongqing, China
| |
Collapse
|
20
|
De Silva P, Saad MA, Thomsen HC, Bano S, Ashraf S, Hasan T. Photodynamic therapy, priming and optical imaging: Potential co-conspirators in treatment design and optimization - a Thomas Dougherty Award for Excellence in PDT paper. J PORPHYR PHTHALOCYA 2020; 24:1320-1360. [PMID: 37425217 PMCID: PMC10327884 DOI: 10.1142/s1088424620300098] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Photodynamic therapy is a photochemistry-based approach, approved for the treatment of several malignant and non-malignant pathologies. It relies on the use of a non-toxic, light activatable chemical, photosensitizer, which preferentially accumulates in tissues/cells and, upon irradiation with the appropriate wavelength of light, confers cytotoxicity by generation of reactive molecular species. The preferential accumulation however is not universal and, depending on the anatomical site, the ratio of tumor to normal tissue may be reversed in favor of normal tissue. Under such circumstances, control of the volume of light illumination provides a second handle of selectivity. Singlet oxygen is the putative favorite reactive molecular species although other entities such as nitric oxide have been credibly implicated. Typically, most photosensitizers in current clinical use have a finite quantum yield of fluorescence which is exploited for surgery guidance and can also be incorporated for monitoring and treatment design. In addition, the photodynamic process alters the cellular, stromal, and/or vascular microenvironment transiently in a process termed photodynamic priming, making it more receptive to subsequent additional therapies including chemo- and immunotherapy. Thus, photodynamic priming may be considered as an enabling technology for the more commonly used frontline treatments. Recently, there has been an increase in the exploitation of the theranostic potential of photodynamic therapy in different preclinical and clinical settings with the use of new photosensitizer formulations and combinatorial therapeutic options. The emergence of nanomedicine has further added to the repertoire of photodynamic therapy's potential and the convergence and co-evolution of these two exciting tools is expected to push the barriers of smart therapies, where such optical approaches might have a special niche. This review provides a perspective on current status of photodynamic therapy in anti-cancer and anti-microbial therapies and it suggests how evolving technologies combined with photochemically-initiated molecular processes may be exploited to become co-conspirators in optimization of treatment outcomes. We also project, at least for the short term, the direction that this modality may be taking in the near future.
Collapse
Affiliation(s)
- Pushpamali De Silva
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Mohammad A. Saad
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Hanna C. Thomsen
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Shazia Bano
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Shoaib Ashraf
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
21
|
Kaneko K, Osada T, Morse MA, Gwin WR, Ginzel JD, Snyder JC, Yang XY, Liu CX, Diniz MA, Bodoor K, Hughes PF, Haystead TA, Lyerly HK. Heat shock protein 90-targeted photodynamic therapy enables treatment of subcutaneous and visceral tumors. Commun Biol 2020; 3:226. [PMID: 32385408 PMCID: PMC7210113 DOI: 10.1038/s42003-020-0956-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 04/21/2020] [Indexed: 01/10/2023] Open
Abstract
Photodynamic therapy (PDT) ablates malignancies by applying focused near-infrared (nIR) light onto a lesion of interest after systemic administration of a photosensitizer (PS); however, the accumulation of existing PS is not tumor-exclusive. We developed a tumor-localizing strategy for PDT, exploiting the high expression of heat shock protein 90 (Hsp90) in cancer cells to retain high concentrations of PS by tethering a small molecule Hsp90 inhibitor to a PS (verteporfin, VP) to create an Hsp90-targeted PS (HS201). HS201 accumulates to a greater extent than VP in breast cancer cells both in vitro and in vivo, resulting in increased treatment efficacy of HS201-PDT in various human breast cancer xenografts regardless of molecular and clinical subtypes. The therapeutic index achieved with Hsp90-targeted PDT would permit treatment not only of localized tumors, but also more diffusely infiltrating processes such as inflammatory breast cancer.
Collapse
Affiliation(s)
- Kensuke Kaneko
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA
| | - Takuya Osada
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA
| | - Michael A Morse
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA.,Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - William R Gwin
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Joshua D Ginzel
- Department of Cell Biology, Duke University, Durham, NC, 27710, USA
| | - Joshua C Snyder
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA.,Department of Cell Biology, Duke University, Durham, NC, 27710, USA
| | - Xiao-Yi Yang
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA
| | - Cong-Xiao Liu
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA
| | - Márcio A Diniz
- Biostatistics and Bioinformatics Research Center, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Khaldon Bodoor
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Philip F Hughes
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Timothy Aj Haystead
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA.
| | - H Kim Lyerly
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA.
| |
Collapse
|
22
|
Cui X, Zhu J, Wu X, Yang S, Yao X, Zhu W, Xu P, Chen X. Hematoporphyrin monomethyl ether-mediated photodynamic therapy inhibits the growth of keloid graft by promoting fibroblast apoptosis and reducing vessel formation. Photochem Photobiol Sci 2020; 19:114-125. [PMID: 31934700 DOI: 10.1039/c9pp00311h] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Photodynamic therapy (PDT) has been shown to significantly inhibit fibroblast activity. However, the effect of PDT mediated by the photosensitizer hematoporphyrin monomethyl ether (HMME) on keloids is not known well. The aim of our study was to examine the efficacy of HMME-PDT in cellular and animal models of keloids. Keloid fibroblasts (KFbs) were isolated from human keloid specimens and the proliferation, invasion, and migration of KFbs after HMME-PDT treatment was examined in vitro. Apoptosis in cells was measured by flow cytometry. Cysteinyl aspartate specific proteinase 3 (Caspase3) expression was determined by immunofluorescence staining and western blot. HMME-PDT inhibited KFbs proliferation, invasion, migration, increased apoptosis rate and enhanced caspase3 and cleaved caspase3 expression. The keloid graft transplantation was performed by using nude mice. The growth of the graft was monitored every third day. Interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) mRNA expression were detected by quantitative real time PCR. It was observed that HMME-PDT attenuated graft growth and reduced vessel density in the keloid grafts. However, HMME-PDT did not alter IL-6 and TNF-α mRNA expression in the keloid grafts. Moreover, HMME-PDT suppressed transforming growth-β1 (TGF-β1) and small phenotype and Drosophila Mothers Against Decapentaplegic 3 (Smad3) expression in both KFbs and keloid grafts. Collectively, the evidence suggests that HMME-PDT inhibits the growth of the keloid graft by promoting the apoptosis of fibroblasts and reducing vessel formation of the keloid graft.
Collapse
Affiliation(s)
- Xiaomei Cui
- Department of Medical Cosmetology, Department of Dermatology, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Jing Zhu
- Department of Dermatology, Wuxi People's Hospital, Wuxi, 214000, China
| | - Xiaoyan Wu
- Department of Medical Cosmetology, Department of Dermatology, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Shengju Yang
- Department of Medical Cosmetology, Department of Dermatology, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Xiaodong Yao
- Department of Medical Cosmetology, Department of Dermatology, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Wenyan Zhu
- Department of Medical Cosmetology, Department of Dermatology, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Pan Xu
- Department of Medical Cosmetology, Department of Dermatology, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Xiaodong Chen
- Department of Medical Cosmetology, Department of Dermatology, Affiliated Hospital of Nantong University, Nantong, 226001, China.
| |
Collapse
|
23
|
Kang MWC, Liu H, Kah JCY. Innate immune activation by conditioned medium of cancer cells following combined phototherapy with photosensitizer-loaded gold nanorods. J Mater Chem B 2020; 8:10812-10824. [DOI: 10.1039/d0tb01953d] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nanoparticle-based phototherapy has evolved to include immunotherapy as an effective treatment combination for cancers through inducing anti-cancer immune activation leading to downstream adaptive responses and immune protection.
Collapse
Affiliation(s)
- Malvin Wei Cherng Kang
- NUS Graduate School for Integrative Sciences and Engineering
- National University of Singapore
- Singapore 119077
- Singapore
| | - Haiyan Liu
- NUS Graduate School for Integrative Sciences and Engineering
- National University of Singapore
- Singapore 119077
- Singapore
- Deparment of Microbiology & Immunology
| | - James Chen Yong Kah
- NUS Graduate School for Integrative Sciences and Engineering
- National University of Singapore
- Singapore 119077
- Singapore
- Department of Biomedical Engineering
| |
Collapse
|
24
|
Pucelik B, Arnaut LG, Dąbrowski JM. Lipophilicity of Bacteriochlorin-Based Photosensitizers as a Determinant for PDT Optimization through the Modulation of the Inflammatory Mediators. J Clin Med 2019; 9:E8. [PMID: 31861531 PMCID: PMC7019385 DOI: 10.3390/jcm9010008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 12/14/2019] [Accepted: 12/18/2019] [Indexed: 12/20/2022] Open
Abstract
: Photodynamic therapy (PDT) augments the host antitumor immune response, but the role of the PDT effect on the tumor microenvironment in dependence on the type of photosensitizer and/or therapeutic protocols has not been clearly elucidated. We employed three bacteriochlorins (F2BOH, F2BMet and Cl2BHep) of different polarity that absorb near-infrared light (NIR) and generated a large amount of reactive oxygen species (ROS) to compare the PDT efficacy after various drug-to-light intervals: 15 min. (V-PDT), 3h (E-PDT) and 72h (C-PDT). We also performed the analysis of the molecular mechanisms of PDT crucial for the generation of the long-lasting antitumor immune response. PDT-induced damage affected the integrity of the host tissue and developed acute (protocol-dependent) local inflammation, which in turn led to the infiltration of neutrophils and macrophages. In order to further confirm this hypothesis, a number of proteins in the plasma of PDT-treated mice were identified. Among a wide range of cytokines (IL-6, IL-10, IL-13, IL-15, TNF-α, GM-CSF), chemokines (KC, MCP-1, MIP1α, MIP1β, MIP2) and growth factors (VEGF) released after PDT, an important role was assigned to IL-6. PDT protocols optimized for studied bacteriochlorins led to a significant increase in the survival rate of BALB/c mice bearing CT26 tumors, but each photosensitizer (PS) was more or less potent, depending on the applied DLI (15 min, 3 h or 72 h). Hydrophilic (F2BOH) and amphiphilic (F2BMet) PSs were equally effective in V-PDT (>80 cure rate). F2BMet was the most efficient in E-PDT (DLI = 3h), leading to a cure of 65 % of the animals. Finally, the most powerful PS in the C-PDT (DLI = 72 h) regimen turned out to be the most hydrophobic compound (Cl2BHep), allowing 100 % of treated animals to be cured at a light dose of only 45 J/cm2.
Collapse
Affiliation(s)
- Barbara Pucelik
- Faculty of Chemistry, Jagiellonian University, 30-387 Kraków, Poland;
- Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Luis G. Arnaut
- CQC, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal;
| | | |
Collapse
|
25
|
Nie S, Wang X, Wang H. NLRP3 Inflammasome Mediated Interleukin-1β Production in Cancer-Associated Fibroblast Contributes to ALA-PDT for Cutaneous Squamous Cell Carcinoma. Cancer Manag Res 2019; 11:10257-10267. [PMID: 31849516 PMCID: PMC6912005 DOI: 10.2147/cmar.s226356] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/11/2019] [Indexed: 12/22/2022] Open
Abstract
Background Long-term tumor control following PDT is a result of its direct effect on tumor and vasculature in combination with induction of inflammatory-reactions upregulating the immune system. When PDT induces necrosis of tumors and vascular system, an immune cascade can be initiated to release all kinds of cytokines including IL1β. This further leads to the activation of inflammatory-cells and hence death of tumor cells. Methods Ultraviolet irradiation was used to induce cSCC mice model, gene chip was used to screen inflammatory cytokines, qPCR, ELISA and implanted tumor mice model were used to verify the changes and important role of interleukin-1β, and WB preliminarily explored the production mechanism of interleukin-1β. Results Inflammatory cytokines and receptors transcript screening identify IL1r1 as the top4. After ALA-PDT, IL1r1 and IL1β increased in patients' biopsies, principally in mesenchymal cells. In vivo, the inhibition of ALA-PDT on tumor growth of cutaneous squamous cell carcinoma (cSCC) mice in the group with intralesional injection of anti-IL1β mAb or caspase1-inhibitor was significantly weaker than the control groups. Furthermore, NLRP3-inflammasome and p-p65/p65 were elevated after ALA-PDT mediated IL1β production in cancer-associated-fibroblasts. Discussion By means of activating NLRP3-inflammasome with IL1β production in CAFs, PDT stimulates local acute-inflammatory-response, which further promotes PDT effect for cSCC.
Collapse
Affiliation(s)
- Shu Nie
- Department of Dermatology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, People's Republic of China
| | - Xiuli Wang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, People's Republic of China
| | - Hongwei Wang
- Department of Dermatology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, People's Republic of China
| |
Collapse
|
26
|
He X, Situ B, Gao M, Guan S, He B, Ge X, Li S, Tao M, Zou H, Tang BZ, Zheng L. Stereotactic Photodynamic Therapy Using a Two-Photon AIE Photosensitizer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1905080. [PMID: 31721436 DOI: 10.1002/smll.201905080] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/16/2019] [Indexed: 06/10/2023]
Abstract
Two-photon photodynamic therapy (TP-PDT) is emerging as a powerful strategy for stereotactic targeting of diseased areas, but ideal photosensitizers (PSs) are currently lacking. This work reports a smart PS with aggregation-induced emission (AIE) feature, namely DPASP, for TP-PDT with excellent performances. DPASP exhibits high affinity to mitochondria, superior photostability, large two-photon absorption cross section as well as efficient reactive oxygen species generation, enabling it to achieve photosensitization both in vitro and in vivo under two-photon excitation. Moreover, its capability of stereotactic ablation of targeted cells with high-precision is also successfully demonstrated. All these merits make DPASP a promising TP-PDT candidate for accurate ablation of abnormal tissues with minimal damages to surrounding areas in the treatment of various diseases.
Collapse
Affiliation(s)
- Xiaojing He
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Bo Situ
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Meng Gao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Shujuan Guan
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Bairong He
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoxue Ge
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shiwu Li
- Center for Aggregation-Induced Emission, State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Maliang Tao
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hang Zou
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ben Zhong Tang
- Center for Aggregation-Induced Emission, State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou, 510640, China
- Department of Chemistry and Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science & Technology, Clear Water Bay, Kowloon, Hong Kong, China
- HKUST-Shenzhen Research Institute, Shenzhen, 518057, China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
27
|
Belashov AV, Zhikhoreva AA, Belyaeva TN, Nikolsky NN, Semenova IV, Kornilova ES, Vasyutinskii OS. Quantitative assessment of changes in cellular morphology at photodynamic treatment in vitro by means of digital holographic microscopy. BIOMEDICAL OPTICS EXPRESS 2019; 10:4975-4986. [PMID: 31646023 PMCID: PMC6788599 DOI: 10.1364/boe.10.004975] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/15/2019] [Accepted: 08/29/2019] [Indexed: 05/03/2023]
Abstract
Temporal dependence of changes in the morphological characteristics of cells of two cultured lines of cancer origin, HeLa and A549, induced by photodynamic treatment with Radachlorin photosensitizer, have been monitored using digital holographic microscopy during first two hours after short-term irradiation. The observed post-treatment early dynamics of the phase shift in the transmitted wavefront indicated several distinct scenarios of cell behavior depending upon the irradiation dose. In particular the phase shift increased at low doses, which can be associated with apoptosis, while at high doses it decreased, which can be associated with necrosis. As shown, the two cell types responded differently to similar irradiation doses. Although the sequence of death scenarios with the increase of the irradiation dose was the same, each scenario was realized at substantially different doses. These findings suggest that the average phase shift of the transmitted wavefront can be used for quantitative non-invasive cell death characterization. The conclusions made were cofirmed by commonly used test assays using confocal fluorescent microscopy.
Collapse
Affiliation(s)
- A. V. Belashov
- Ioffe Institute; 26, Polytekhnicheskaya, St. Petersburg, 194021, Russia
| | - A. A. Zhikhoreva
- Ioffe Institute; 26, Polytekhnicheskaya, St. Petersburg, 194021, Russia
| | - T. N. Belyaeva
- Institute of Cytology of RAS; 4, Tikhoretsky pr., St. Petersburg, 194064, Russia
| | - N. N. Nikolsky
- Institute of Cytology of RAS; 4, Tikhoretsky pr., St. Petersburg, 194064, Russia
| | - I. V. Semenova
- Ioffe Institute; 26, Polytekhnicheskaya, St. Petersburg, 194021, Russia
| | - E. S. Kornilova
- Institute of Cytology of RAS; 4, Tikhoretsky pr., St. Petersburg, 194064, Russia
- Peter the Great St. Petersburg Polytechnic University, 29 Polytekhnicheskaya, St. Petersburg, 195251, Russia
- St. Petersburg State University, 7/9, University Embankment, St. Petersburg, 199034, Russia
| | | |
Collapse
|
28
|
Udartseva OO, Zhidkova OV, Ezdakova MI, Ogneva IV, Andreeva ER, Buravkova LB, Gollnick SO. Low-dose photodynamic therapy promotes angiogenic potential and increases immunogenicity of human mesenchymal stromal cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 199:111596. [DOI: 10.1016/j.jphotobiol.2019.111596] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/23/2019] [Accepted: 08/14/2019] [Indexed: 12/19/2022]
|
29
|
Hameed S, Mo S, Mustafa G, Bajwa SZ, Khan WS, Dai Z. Immunological Consequences of Nanoparticle‐Mediated Antitumor Photoimmunotherapy. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900101] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Sadaf Hameed
- Department of Biomedical EngineeringCollege of EngineeringPeking University Beijing 100871 China
| | - Shanyan Mo
- Department of Biomedical EngineeringCollege of EngineeringPeking University Beijing 100871 China
| | - Ghulam Mustafa
- Department of SciencesBahria University Lahore Lahore 54000 Pakistan
| | - Sadia Z. Bajwa
- Nanobiotech GroupNational Institute for Biotechnology and Genetic Engineering (NIBGE) P.O. Box No. 577, Jhang Road Faisalabad 44000 Pakistan
| | - Waheed S. Khan
- Nanobiotech GroupNational Institute for Biotechnology and Genetic Engineering (NIBGE) P.O. Box No. 577, Jhang Road Faisalabad 44000 Pakistan
| | - Zhifei Dai
- Department of Biomedical EngineeringCollege of EngineeringPeking University Beijing 100871 China
| |
Collapse
|
30
|
Voisin M, Nourshargh S. Neutrophil trafficking to lymphoid tissues: physiological and pathological implications. J Pathol 2019; 247:662-671. [PMID: 30584795 PMCID: PMC6492258 DOI: 10.1002/path.5227] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 12/20/2018] [Accepted: 12/23/2018] [Indexed: 12/11/2022]
Abstract
Recent advances have provided evidence for the involvement of neutrophils in both innate and adaptive immunity, robustly challenging the old dogma that neutrophils are short-lived prototypical innate immune cells solely involved in acute responses to microbes and exerting collateral tissue damage. There is now ample evidence showing that neutrophils can migrate into different compartments of the lymphoid system where they contribute to the orchestration of the activation and/or suppression of lymphocyte effector functions in homeostasis and during chronic inflammation, such as autoimmune disorders and cancer. In support of this notion, neutrophils can generate a wide range of cytokines and other mediators capable of regulating the survival, proliferation and functions of both T and B cells. In addition, neutrophils can directly engage with lymphocytes and promote antigen presentation. Furthermore, there is emerging evidence of the existence of distinct and diverse neutrophil phenotypes with immunomodulatory functions that characterise different pathological conditions, including chronic and autoimmune inflammatory conditions. The aim of this review is to discuss the mechanisms implicated in neutrophil trafficking into the lymphoid system and to provide an overview of the immuno-regulatory functions of neutrophils in health and disease in the context of adaptive immunity. Copyright © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Mathieu‐Benoit Voisin
- Centre for Microvascular Research, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of LondonLondonUK
| | - Sussan Nourshargh
- Centre for Microvascular Research, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of LondonLondonUK
| |
Collapse
|
31
|
Anand S, Yasinchak A, Govande M, Shakya S, Maytin EV. Painless versus conventional photodynamic therapy for treatment of actinic keratosis: Comparison of cell death and immune response in a murine model. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2019; 10860:108600K. [PMID: 33790491 PMCID: PMC8009283 DOI: 10.1117/12.2511646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Aminolevulinic acid based photodynamic therapy (ALA-PDT) is a popular and efficacious treatment for actinic keratosis (AK). However, standard PDT can elicit stinging pain during illumination, and hence is not always favored by patients. In a new regimen called metronomic PDT (mPDT), similar to daylight PDT but using blue light, the illumination is delivered concurrently with ALA application rather than after a 1-hour pre-incubation (conventional regimen, cPDT). In the clinic, mPDT is not only painless but also nearly as effective as cPDT for AK lesion clearance. In this investigation, a murine AK model (generated by repeated UVB exposure) was treated with either mPDT or cPDT. Lesion clearance was followed by area measurement, and samples were harvested for mechanistic analyses. Compared to pretreatment (100%), the average lesion area was reduced to 47% and 32% in cPDT, and to 57% and 40% in mPDT at 1- and 2-weeks post PDT, respectively. Relative to untreated controls, enhanced cell death (histomorphology by H&E staining and apoptosis by TUNEL assay), and generation of Reactive Oxygen Species (ROS; CM-H2DCFDA staining) were observed in both cPDT and mPDT samples. Activation of cleaved Caspase-3 was specifically observed only in cPDT samples. Immunomodulation by inflammatory cells was observed by enhanced infiltration/retention of neutrophils and macrophages in metronomic PDT samples. Our results suggest that metronomic PDT can be just as effective as conventional PDT for treatment of AK, but the mechanisms may be quite different.
Collapse
Affiliation(s)
- Sanjay Anand
- Department of Biomedical Engineering, Lerner Research Institute
- Dermatology and Plastic Surgery Institute
- Cleveland Clinic Lerner College of Medicine of CWRU, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Anton Yasinchak
- Department of Biomedical Engineering, Lerner Research Institute
| | - Mukul Govande
- Department of Biomedical Engineering, Lerner Research Institute
| | - Sajina Shakya
- Department of Biomedical Engineering, Lerner Research Institute
| | - Edward V. Maytin
- Department of Biomedical Engineering, Lerner Research Institute
- Dermatology and Plastic Surgery Institute
- Cleveland Clinic Lerner College of Medicine of CWRU, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| |
Collapse
|
32
|
Li Y, Li X, Zhou F, Doughty A, Hoover AR, Nordquist RE, Chen WR. Nanotechnology-based photoimmunological therapies for cancer. Cancer Lett 2019; 442:429-438. [PMID: 30476523 PMCID: PMC6613935 DOI: 10.1016/j.canlet.2018.10.044] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/24/2018] [Accepted: 10/25/2018] [Indexed: 12/16/2022]
Abstract
Phototherapy is a non-invasive or minimally invasive therapeutic strategy. Immunotherapy uses different immunological approaches, such as antibodies, vaccines, immunoadjuvants, and cytokines to stimulate the host immune system to fight against diseases. In cancer treatment, phototherapy not only destroys tumor cells, but also induces immunogenic tumor cell death to initiate a systemic anti-tumor immune response. When combined with immunotherapy, the effectiveness of phototherapy can be enhanced. Because of their special physical, chemical, and sometimes immunological properties, nanomaterials have also been used to enhance phototherapy. In this article, we review the recent progress in nanotechnology-based phototherapy, including nano-photothermal therapy, nano-photochemical therapy, and nano-photoimmunological therapy in cancer treatment. Specifically, we focus on the immunological responses induced by nano-phototherapies.
Collapse
Affiliation(s)
- Yong Li
- Interventional Therapy Department, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China; Biophotonics Research Laboratory, Center for Interdisciplinary Biomedical Education and Research, College of Mathematics and Science, University of Central Oklahoma, Edmond, OK, 73034, USA
| | - Xiaosong Li
- Department of Oncology, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing 100048, China
| | - Feifan Zhou
- Biophotonics Research Laboratory, Center for Interdisciplinary Biomedical Education and Research, College of Mathematics and Science, University of Central Oklahoma, Edmond, OK, 73034, USA; Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education/Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, PR China
| | - Austin Doughty
- Biophotonics Research Laboratory, Center for Interdisciplinary Biomedical Education and Research, College of Mathematics and Science, University of Central Oklahoma, Edmond, OK, 73034, USA
| | - Ashley R Hoover
- Biophotonics Research Laboratory, Center for Interdisciplinary Biomedical Education and Research, College of Mathematics and Science, University of Central Oklahoma, Edmond, OK, 73034, USA
| | - Robert E Nordquist
- Immunophotonics Inc., 4320 Forest Park Avenue #303, St. Louis, Missouri 63108, USA
| | - Wei R Chen
- Biophotonics Research Laboratory, Center for Interdisciplinary Biomedical Education and Research, College of Mathematics and Science, University of Central Oklahoma, Edmond, OK, 73034, USA; Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education/Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, PR China.
| |
Collapse
|
33
|
Rice SR, Li YR, Busch TM, Kim MM, McNulty S, Dimofte A, Zhu TC, Cengel KA, Simone CB. A Novel Prospective Study Assessing the Combination of Photodynamic Therapy and Proton Radiation Therapy: Safety and Outcomes When Treating Malignant Pleural Mesothelioma. Photochem Photobiol 2019; 95:411-418. [PMID: 30485442 PMCID: PMC6778401 DOI: 10.1111/php.13065] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/20/2018] [Indexed: 12/11/2022]
Abstract
Malignant pleural mesothelioma remains difficult to treat, with high failure rates despite optimal therapy. We present a novel prospective trial combining proton therapy (PT) and photodynamic therapy (PDT) and the largest-ever mesothelioma PT experience (n = 10). PDT photosensitizers included porfimer sodium (2 mg·kg-1 ; 24 h drug-light interval) or 2-[1-hexyloxyethyl]-2-devinyl pyropheophorbide-a (HPPH) (4 mg·m-2 ;48 h) with wavelengths of 630 nm to 60J·cm-2 and 665 nm to 15-45J·cm-2 , respectively. With a median age of 69 years, patients were predominantly male (90%) with epithelioid histology (100%) and stage III-IV disease (100%). PT was delivered to a median of 55.0 CGE/1.8-2.0 CGE (range 50-75 CGE) adjuvantly (n = 8) or as salvage therapy (n = 2) following extended pleurectomy/decortication (ePD)/PDT. Two-year local control was 90%, with distant and regional failure rates of 50% and 30%, respectively. All patients received chemotherapy, and four received immunotherapy. Surgical complications included atrial fibrillation (n = 3), pneumonia (n = 2), and deep vein thrombosis (n = 2). Median survival from PT completion was 19.5 months (30.3 months from diagnosis), and 1- and 2-year survival rates were 58% and 29%. No patient experienced CTCAEv4 grade ≥2 acute or late toxicity. Our prolonged survival in very advanced-stage patients compares favorably to survival for PT without PDT and photon therapy with PDT, suggesting possible spatial or systemic cooperativity and immune effect.
Collapse
Affiliation(s)
- Stephanie R. Rice
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD
| | - Yun R. Li
- Helen Diller Family Comprehensive Cancer Center, Department of Radiation Oncology, University of California San Francisco, San Francisco, CA
| | - Theresa M. Busch
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Michele M. Kim
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Sally McNulty
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Andrea Dimofte
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Timothy C. Zhu
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Keith A. Cengel
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Charles B. Simone
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
34
|
Davis RW, Snyder E, Miller J, Carter S, Houser C, Klampatsa A, Albelda SM, Cengel KA, Busch TM. Luminol Chemiluminescence Reports Photodynamic Therapy-Generated Neutrophil Activity In Vivo and Serves as a Biomarker of Therapeutic Efficacy. Photochem Photobiol 2018; 95:430-438. [PMID: 30357853 DOI: 10.1111/php.13040] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 10/03/2018] [Indexed: 01/26/2023]
Abstract
Inflammatory cells, most especially neutrophils, can be a necessary component of the antitumor activity occurring after administration of photodynamic therapy. Generation of neutrophil responses has been suggested to be particularly important in instances when the delivered photodynamic therapy (PDT) dose is insufficient. In these cases, the release of neutrophil granules and engagement of antitumor immunity may play an important role in eliminating residual disease. Herein, we utilize in vivo imaging of luminol chemiluminescence to noninvasively monitor neutrophil activation after PDT administration. Studies were performed in the AB12 murine model of mesothelioma, treated with Photofrin-PDT. Luminol-generated chemiluminescence increased transiently 1 h after PDT, followed by a subsequent decrease at 4 h after PDT. The production of luminol signal was not associated with the influx of Ly6G+ cells, but was related to oxidative burst, as an indicator of neutrophil function. Most importantly, greater levels of luminol chemiluminescence 1 h after PDT were prognostic of a complete response at 90 days after PDT. Taken together, this research supports an important role for early activity by Ly6G+ cells in the generation of long-term PDT responses in mesothelioma, and it points to luminol chemiluminescence as a potentially useful approach for preclinical monitoring of neutrophil activation by PDT.
Collapse
Affiliation(s)
- Richard W Davis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Emma Snyder
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Joann Miller
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Shirron Carter
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Cassandra Houser
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Astero Klampatsa
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Steven M Albelda
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Keith A Cengel
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Theresa M Busch
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
35
|
Dadar M, Chakraborty S, Dhama K, Prasad M, Khandia R, Hassan S, Munjal A, Tiwari R, Karthik K, Kumar D, Iqbal HMN, Chaicumpa W. Advances in Designing and Developing Vaccines, Drugs and Therapeutic Approaches to Counter Human Papilloma Virus. Front Immunol 2018; 9:2478. [PMID: 30483247 PMCID: PMC6240620 DOI: 10.3389/fimmu.2018.02478] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 10/08/2018] [Indexed: 02/05/2023] Open
Abstract
Human papillomavirus (HPV) is a viral infection with skin-to-skin based transmission mode. HPV annually caused over 500,000 cancer cases including cervical, anogenital and oropharyngeal cancer among others. HPV vaccination has become a public-health concern, worldwide, to prevent the cases of HPV infections including precancerous lesions, cervical cancers, and genital warts especially in adolescent female and male population by launching national programs with international alliances. Currently, available prophylactic and therapeutic vaccines are expensive to be used in developing countries for vaccination programs. The recent progress in immunotherapy, biotechnology, recombinant DNA technology and molecular biology along with alternative and complementary medicinal systems have paved novel ways and valuable opportunities to design and develop effective prophylactic and therapeutic vaccines, drugs and treatment approach to counter HPV effectively. Exploration and more researches on such advances could result in the gradual reduction in the incidences of HPV cases across the world. The present review presents a current global scenario and futuristic prospects of the advanced prophylactic and therapeutic approaches against HPV along with recent patents coverage of the progress and advances in drugs, vaccines and therapeutic regimens to effectively combat HPV infections and its cancerous conditions.
Collapse
Affiliation(s)
- Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization, Karaj, Iran
| | - Sandip Chakraborty
- Department of Veterinary Microbiology, College of Veterinary Sciences and Animal Husbandry, West Tripura, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Minakshi Prasad
- Department of Animal Biotechnology, LLR University of Veterinary and Animal Sciences, Hisar, India
| | - Rekha Khandia
- Department of Genetics, Barkatullah University, Bhopal, India
| | - Sameer Hassan
- Department of Biomedical Informatics, National Institute for Research in Tuberculosis, Indian Council of Medical Research, Chennai, India
| | - Ashok Munjal
- Department of Genetics, Barkatullah University, Bhopal, India
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, U P Pt. Deen Dayal Upadhayay Pashu Chikitsa Vigyan Vishwavidyalay Evum Go-Anusandhan Sansthan, Mathura, India
| | - Kumaragurubaran Karthik
- Central University Laboratory, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | - Deepak Kumar
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Hafiz M. N. Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey, Mexico
| | - Wanpen Chaicumpa
- Department of Parasitology, Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Faculty of Medicine SIriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
36
|
Hwang HS, Shin H, Han J, Na K. Combination of photodynamic therapy (PDT) and anti-tumor immunity in cancer therapy. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2018; 48:143-151. [PMID: 30680248 PMCID: PMC6323106 DOI: 10.1007/s40005-017-0377-x] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 12/02/2017] [Indexed: 01/10/2023]
Abstract
Photodynamic therapy (PDT) is performed using a photosensitizer and light of specific wavelength in the presence of oxygen to generate singlet oxygen and reactive oxygen species(ROS) in the cancer cells. The accumulated photosensitizers in target sites induce ROS generation upon light activation, then the generated cytotoxic reactive oxygen species lead to tumor cell death via apoptosis or necrosis, and damages the target sites which results tumor destruction. As a consequence, the PDT-mediated cell death is associated with anti-tumor immune response. In this paper, the effects of PDT and immune response on tumors are reviewed. Activation of an immune response regarding the innate and adaptive immune response, interaction with immune cells and tumor cells that associated with antitumor efficacy of PDT are also discussed.
Collapse
Affiliation(s)
- Hee Sook Hwang
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonni-gu, Bucheno-si, Gyeonggido 14662 South Korea
| | - Heejun Shin
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonni-gu, Bucheno-si, Gyeonggido 14662 South Korea
| | - Jieun Han
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonni-gu, Bucheno-si, Gyeonggido 14662 South Korea
| | - Kun Na
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonni-gu, Bucheno-si, Gyeonggido 14662 South Korea
| |
Collapse
|
37
|
Abstract
Photodynamic therapy of tumors requires the topical, systemic or oral administration of a photosensitizing compound, illumination of the tumor area by light of a specific wavelength and the presence of oxygen. Light activation of the photosensitizer transfers energy to molecular oxygen creating singlet oxygen, a highly reactive and toxic species that rapidly reacts with cellular components causing oxidative damage, ultimately leading to cell death. Tumor destruction caused by photodynamic therapy is not only a result of direct tumor cell toxicity via the generation of reactive oxygen species but there is also an immunological and vascular component involved. The immune response to photodynamic therapy has been demonstrated to significantly enhance its efficacy. Depending on a number of factors, including type of photosensitizer, light dose and dose rate, photodynamic therapy has been shown to induce cell death via apoptosis, necrosis, autophagy and in particular immunogenic cell death. It is the purpose of this review to focus mainly on the role photodynamic therapy could play in the generation of specific anti-tumor immunity and vaccines for the treatment of brain tumors.
Collapse
Affiliation(s)
- Henry Hirschberg
- Beckman Laser Institute and Medical Clinic, University of California, Irvine, CA 92617, USA
| | - Kristian Berg
- Department of Radiation Biology, Norwegian Radium Hospital, Oslo University Hospital, Montebello, Oslo N-0310, Norway
| | - Qian Peng
- Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, Montebello, Oslo N-0310, Norway
| |
Collapse
|
38
|
Reboredo-Rodríguez P, González-Barreiro C, Cancho-Grande B, Simal-Gándara J, Giampieri F, Forbes-Hernández TY, Gasparrini M, Afrin S, Cianciosi D, Manna PP, Varela-López A, Ojeda-Amador RM, Fregapane G, Desamparados Salvador M, Battino M. Effect of pistachio kernel extracts in MCF-7 breast cancer cells: Inhibition of cell proliferation, induction of ROS production, modulation of glycolysis and of mitochondrial respiration. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.03.045] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
39
|
Davis RW, Papasavvas E, Klampatsa A, Putt M, Montaner LJ, Culligan MJ, McNulty S, Friedberg JS, Simone CB, Singhal S, Albelda SM, Cengel KA, Busch TM. A preclinical model to investigate the role of surgically-induced inflammation in tumor responses to intraoperative photodynamic therapy. Lasers Surg Med 2018; 50:440-450. [PMID: 29799130 DOI: 10.1002/lsm.22934] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2018] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Inflammation is a well-known consequence of surgery. Although surgical debulking of tumor is beneficial to patients, the onset of inflammation in injured tissue may impede the success of adjuvant therapies. One marker for postoperative inflammation is IL-6, which is released as a consequence of surgical injuries. IL-6 is predictive of response to many cancer therapies, and it is linked to various molecular and cellular resistance mechanisms. The purpose of this study was to establish a murine model by which therapeutic responses to photodynamic therapy (PDT) can be studied in the context of surgical inflammation. MATERIALS AND METHODS Murine models with AB12 mesothelioma tumors were treated with either surgical resection or sham surgery with tumor incision but no resection. The timing and extent of IL-6 release in the tumor and/or serum was measured using enzyme-linked immunosorbent assay (ELISA) and compared to that measured in the serum of 27 consecutive, prospectively enrolled patients with malignant pleural mesothelioma (MPM) who underwent macroscopic complete resection (MCR). RESULTS MPM patients showed a significant increase in IL-6 at the time MCR was completed. Similarly, IL-6 increased in the tumor and serum of mice treated with surgical resections. However, investigations that combine resection with another therapy make it necessary to grow tumors for resection to a larger volume than those that receive secondary therapy alone. As the larger size may alter tumor biology independent of the effects of surgical injury, we assessed the tumor incision model. In this model, tumor levels of IL-6 significantly increased after tumor incision. CONCLUSION The tumor incision model induces IL-6 release as is seen in the surgical setting, yet it avoids the limitations of surgical resection models. Potential mechanisms by which surgical induction of inflammation and IL-6 could alter the nature and efficacy of tumor response to PDT are reviewed. These include a wide spectrum of molecular and cellular mechanisms through which surgically-induced IL-6 could change the effectiveness of therapies that are combined with surgery. The tumor incision model can be employed for novel investigations of the effects of surgically-induced, acute inflammation on therapeutic response to PDT (or potentially other therapies). Lasers Surg. Med. 50:440-450, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Richard W Davis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | | | - Astero Klampatsa
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Mary Putt
- Department of Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Luis J Montaner
- Wistar Institute, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Melissa J Culligan
- Division of Thoracic Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Sally McNulty
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Joseph S Friedberg
- Division of Thoracic Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Charles B Simone
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Sunil Singhal
- Division of Thoracic Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Steven M Albelda
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Keith A Cengel
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | - Theresa M Busch
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| |
Collapse
|
40
|
Srivatsan A, Sen A, Cheruku RR, Missert JR, Durrani FA, Guru K, Pandey RK. Whole body and local hyperthermia enhances the photosensitizing efficacy of 3-[(1'-hexyloxy)ethyl]-3-Devinylpyropheophorbide-a (HPPH). Lasers Surg Med 2018; 50:506-512. [PMID: 29737551 DOI: 10.1002/lsm.22932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2018] [Indexed: 11/11/2022]
Abstract
BACKGROUND AND OBJECTIVES In this study, we evaluated the impact of hyperthermia in photosensitizing efficacy of 3-[(1'-hexyloxy)ethyl-3-devinylpyropheophorbide-a (HPPH or Photochlor) for the treatment of cancer by photodynamic therapy (PDT). STUDY DESIGN/MATERIALS AND METHODS The outcome of both whole body hyperthermia (WBH) and local hyperthermia (LH) in combination with HPPH-PDT was determined in BALB/c and nude mice bearing Colon26 and U87 tumors, respectively. LH was performed by using an indigenously designed heating device, that was heated to the required temperature using a circulating water bath. The device which has flexible membrane on one side was placed on skin above the tumor. The temperature of the tumor was monitored using a thermocouple sensor placed on the surface of the tumor capable of measuring the temperature within 0.1°C. Uptake of the photosensitizer in tumors was determined by fluorescence using an IVIS or a Nuance Imaging System. The PDT was performed by exposing the tumors to 665 nm laser loght, (135 J/cm2 , 75 mW/cm2 ) at the maximal uptake time of HPPH. Tumor size was measured daily using vernier calipers. RESULTS The improved PDT efficacy (long-term percentage tumor cure) in combination with hyperthermia is possible due to an increase in tumor-uptake of the photosensitizer (PS), confirmed by in vivo fluorescence imaging and also by increased tumor perfusion and decreased hypoxia as have been reported previously (Sen et al. [2011] Cancer Res. 71:3872-3880 In Vivo. 20:689-695). Interestingly, compared to whole body hyperthermia, the 14 C- HPPH biodistribution data under local hyperthermia showed similar tumor-uptake in BALB/c mice bearing Colon26 tumors, but significantly lower uptake in other organs and in the blood. CONCLUSION Our study demonstrates that both, fever range whole body and local hyperthermia in combination with HPPH-PDT enhances the long-term tumor cure of BALB/c and nude mice implanted with Colon26 and U87 tumors respectively. Lasers Surg. Med. 50:506-512, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Avinash Srivatsan
- PDT Center, Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, 14263
| | - Arindam Sen
- PDT Center, Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, 14263
| | - Ravindra R Cheruku
- PDT Center, Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, 14263
| | - Joseph R Missert
- PDT Center, Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, 14263
| | - Farukh A Durrani
- PDT Center, Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, 14263
| | - Khurshid Guru
- Department of Urology, Roswell Park Cancer Institute, Buffalo, New York, 14263
| | - Ravindra K Pandey
- PDT Center, Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, 14263
| |
Collapse
|
41
|
Khan TK, Wender PA, Alkon DL. Bryostatin and its synthetic analog, picolog rescue dermal fibroblasts from prolonged stress and contribute to survival and rejuvenation of human skin equivalents. J Cell Physiol 2018; 233:1523-1534. [PMID: 28590053 PMCID: PMC5673504 DOI: 10.1002/jcp.26043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 06/06/2017] [Indexed: 11/09/2022]
Abstract
Skin health is associated with the day-to-day activity of fibroblasts. The primary function of fibroblasts is to synthesize structural proteins, such as collagen, extracellular matrix proteins, and other proteins that support the structural integrity of the skin and are associated with younger, firmer, and more elastic skin that is better able to resist and recover from injury. At sub-nanomolar concentrations (0.03-0.3 nM), bryostatin-1 and its synthetic analog, picolog (0.1-10 nM) sustained the survival and activation of human dermal fibroblasts cultured under the stressful condition of prolonged serum deprivation. Bryostatin-1 treatment stabilized human skin equivalents (HSEs), a bioengineered combination of primary human skin cells (keratinocytes and dermal fibroblasts) on an extracellular matrix composed of mainly collagen. Fibroblasts activated by bryostatin-1 protected the structural integrity of HSEs. Bryostatin-1 and picolog prolonged activation of Erk in fibroblasts to promote cell survival. Chronic stress promotes the progression of apoptosis. Dermal fibroblasts constitutively express all components of Fas associated apoptosis, including caspase-8, an initiator enzyme of apoptosis. Prolong bryostatin-1 treatment reduced apoptosis by decreasing caspase-8 and protected dermal fibroblasts. Our data suggest that bryostatin-1 and picolog could be useful in anti-aging skincare, and could have applications in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Tapan K. Khan
- Center for Neurodegenerative diseases, Blanchette Rockefeller Neurosciences Institute at West Virginia University, Morgantown, WV 26506, USA
| | - Paul A. Wender
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Daniel L. Alkon
- Neurotrope BioScience, 205 East 42nd Street, 16th Floor, New York, NY 10017, USA
| |
Collapse
|
42
|
Madsen SJ, Christie C, Huynh K, Peng Q, Uzal FA, Krasieva TB, Hirschberg H. Limiting glioma development by photodynamic therapy-generated macrophage vaccine and allo-stimulation: an in vivo histological study in rats. JOURNAL OF BIOMEDICAL OPTICS 2018; 23:1-7. [PMID: 29417766 PMCID: PMC5802332 DOI: 10.1117/1.jbo.23.2.028001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 01/19/2018] [Indexed: 06/08/2023]
Abstract
Immunotherapy of brain tumors involves the stimulation of an antitumor immune response. This type of therapy can be targeted specifically to tumor cells thus sparing surrounding normal brain. Due to the presence of the blood-brain barrier, the brain is relatively isolated from the systemic circulation and, as such, the initiation of significant immune responses is more limited than other types of cancers. The purpose of this study was to show that the efficacy of tumor primed antigen presenting macrophage (MaF98) vaccines can be increased by: (1) photodynamic therapy (PDT) of the priming tumor cells and (2) intracranial injection of allogeneic glioma cells directly into the tumor site. Experiments were conducted in an in vivo brain tumor development model using Fischer rats and F98 (syngeneic) and BT4C (allogeneic) glioma cells. The results showed that immunization with Ma (acting as antigen-presenting cells), primed with PDT-treated tumor cells (MaF98), significantly slowed but did not prevent the growth of F98-induced tumors in the brain. Complete suppression of tumor development was obtained via MaF98 inoculation combined with direct intracranial injection of allogeneic glioma cells. No deleterious effects were noted in any of the animals during the 14-day observation period.
Collapse
Affiliation(s)
- Steen J. Madsen
- University of Nevada, Department of Health Physics and Diagnostic Sciences, Las Vegas, Nevada, United States
| | - Catherine Christie
- University of California, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| | - Khoi Huynh
- University of California, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| | - Qian Peng
- University of Oslo, Pathology Clinic, Rikshospitalet-Radiumhospitalet HF Medical Center, Montebello, Oslo, Norway
| | - Francisco A. Uzal
- University of California, School of Veterinary Medicine, Davis, San Bernardino, California, United States
| | - Tatiana B. Krasieva
- University of California, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| | - Henry Hirschberg
- University of California, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| |
Collapse
|
43
|
Nesi-Reis V, Lera-Nonose DSSL, Oyama J, Silva-Lalucci MPP, Demarchi IG, Aristides SMA, Teixeira JJV, Silveira TGV, Lonardoni MVC. Contribution of photodynamic therapy in wound healing: A systematic review. Photodiagnosis Photodyn Ther 2017; 21:294-305. [PMID: 29289704 DOI: 10.1016/j.pdpdt.2017.12.015] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 12/12/2017] [Accepted: 12/26/2017] [Indexed: 10/18/2022]
Abstract
OBJECTIVE We researched articles that used photodynamic therapy (PDT) for skin wound healing in humans. METHODS The systematic review was conducted through scientific articles that investigated the action of PDT on wound healing in humans, published from July 2005 to March 2017, in the data bases PubMed and LILACS. RESULTS The main types of wound described in selected articles in this review were chronic ulcer and non-melanoma skin cancer. For accomplishing the PDT, second generation of photosensitizing agents with laser or light emitting diode were used. The studies demonstrated that PDT contribute in several ways to the wound healing process: leading to cellular death; reducing or increasing inflammation; stimulating fibroblasts proliferation and, consequently, of collagen and elastin; raising transforming growth factor beta and metalloproteinases. Based on this, PDT provided good results in wound healing process, acting in several steps and accelerating tissue repair. CONCLUSIONS PDT improved healing in many wound models in humans, revealing itself as a promising therapeutic modality for stimulating wound healing and remodelling.
Collapse
Affiliation(s)
- Vanessa Nesi-Reis
- Graduate Program in Health Sciences, Universidade Estadual de Maringá, Avenida Colombo, 5790, Jardim Universitário, CEP 87020-900, Maringá, Paraná, Brazil.
| | | | - Jully Oyama
- Graduate Program in Bioscience and Physiopathology, Universidade Estadual de Maringá, Avenida Colombo, 5790, Jardim Universitário, CEP 87020-900, Maringá, Paraná, Brazil.
| | - Marielle Priscila Paula Silva-Lalucci
- Graduate Program in Bioscience and Physiopathology, Universidade Estadual de Maringá, Avenida Colombo, 5790, Jardim Universitário, CEP 87020-900, Maringá, Paraná, Brazil.
| | - Izabel Galhardo Demarchi
- Departament of Clinical Analysis and Biomedicine, Universidade Estadual de Maringá, Avenida Colombo, 5790, Jardim Universitário, CEP 87020-900, Maringá, Paraná, Brazil.
| | - Sandra Mara Alessi Aristides
- Departament of Clinical Analysis and Biomedicine, Universidade Estadual de Maringá, Avenida Colombo, 5790, Jardim Universitário, CEP 87020-900, Maringá, Paraná, Brazil.
| | - Jorge Juarez Vieira Teixeira
- Departament of Clinical Analysis and Biomedicine, Universidade Estadual de Maringá, Avenida Colombo, 5790, Jardim Universitário, CEP 87020-900, Maringá, Paraná, Brazil.
| | - Thaís Gomes Verzignassi Silveira
- Departament of Clinical Analysis and Biomedicine, Universidade Estadual de Maringá, Avenida Colombo, 5790, Jardim Universitário, CEP 87020-900, Maringá, Paraná, Brazil.
| | - Maria Valdrinez Campana Lonardoni
- Departament of Clinical Analysis and Biomedicine, Universidade Estadual de Maringá, Avenida Colombo, 5790, Jardim Universitário, CEP 87020-900, Maringá, Paraná, Brazil.
| |
Collapse
|
44
|
Yurt F, Ocakoglu K, Ince M, Colak SG, Er O, Soylu HM, Gunduz C, Biray Avci C, Caliskan Kurt C. Photodynamic therapy and nuclear imaging activities of zinc phthalocyanine-integrated TiO2
nanoparticles in breast and cervical tumors. Chem Biol Drug Des 2017; 91:789-796. [DOI: 10.1111/cbdd.13144] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 09/07/2017] [Accepted: 11/01/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Fatma Yurt
- Department of Nuclear Applications; Institute of Nuclear Science; Ege University; Izmir Turkey
| | - Kasim Ocakoglu
- Advanced Technology Research & Application Center; Mersin University; Mersin Turkey
- Department of Energy Systems Engineering; Faculty of Tarsus Technology; Mersin University; Mersin Turkey
| | - Mine Ince
- Department of Energy Systems Engineering; Faculty of Tarsus Technology; Mersin University; Mersin Turkey
| | | | - Ozge Er
- Department of Nuclear Applications; Institute of Nuclear Science; Ege University; Izmir Turkey
| | - Hale Melis Soylu
- Department of Biomedical Technology; Institute of Science; Ege University; Izmir Turkey
| | - Cumhur Gunduz
- Department of Medical Biology; Faculty of Medicine; Ege University; Izmir Turkey
| | - Cıgır Biray Avci
- Department of Medical Biology; Faculty of Medicine; Ege University; Izmir Turkey
| | - Cansu Caliskan Kurt
- Department of Medical Biology; Faculty of Medicine; Ege University; Izmir Turkey
| |
Collapse
|
45
|
Malatesti N, Munitic I, Jurak I. Porphyrin-based cationic amphiphilic photosensitisers as potential anticancer, antimicrobial and immunosuppressive agents. Biophys Rev 2017; 9:149-168. [PMID: 28510089 PMCID: PMC5425819 DOI: 10.1007/s12551-017-0257-7] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 03/05/2017] [Indexed: 12/15/2022] Open
Abstract
Photodynamic therapy (PDT) combines a photosensitiser, light and molecular oxygen to induce oxidative stress that can be used to kill pathogens, cancer cells and other highly proliferative cells. There is a growing number of clinically approved photosensitisers and applications of PDT, whose main advantages include the possibility of selective targeting, localised action and stimulation of the immune responses. Further improvements and broader use of PDT could be accomplished by designing new photosensitisers with increased selectivity and bioavailability. Porphyrin-based photosensitisers with amphiphilic properties, bearing one or more positive charges, are an effective tool in PDT against cancers, microbial infections and, most recently, autoimmune skin disorders. The aim of the review is to present some of the recent examples of the applications and research that employ this specific group of photosensitisers. Furthermore, we will highlight the link between their structural characteristics and PDT efficiency, which will be helpful as guidelines for rational design and evaluation of new PSs.
Collapse
Affiliation(s)
- Nela Malatesti
- Department of Biotechnology, University of Rijeka, Radmile Matejčić 2, 51000, Rijeka, Croatia.
| | - Ivana Munitic
- Department of Biotechnology, University of Rijeka, Radmile Matejčić 2, 51000, Rijeka, Croatia
| | - Igor Jurak
- Department of Biotechnology, University of Rijeka, Radmile Matejčić 2, 51000, Rijeka, Croatia
| |
Collapse
|
46
|
Photodynamic Therapy of Non-Small Cell Lung Cancer. Narrative Review and Future Directions. Ann Am Thorac Soc 2016; 13:265-75. [PMID: 26646726 DOI: 10.1513/annalsats.201509-650fr] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Photodynamic therapy (PDT) is an established treatment modality for non-small cell lung cancer. Phototoxicity, the primary adverse event, is expected to be minimized with the introduction of new photosensitizers that have shown promising results in phase I and II clinical studies. Early-stage and superficial endobronchial lesions less than 1 cm in thickness can be effectively treated with external light sources. Thicker lesions and peripheral lesions may be amenable to interstitial PDT, where the light is delivered intratumorally. The addition of PDT to standard-of-care surgery and chemotherapy can improve survival and outcomes in patients with pleural disease. Intraoperative PDT has shown promise in the treatment of non-small cell lung cancer with pleural spread. Recent preclinical and clinical data suggest that PDT can increase antitumor immunity. Crosslinking of signal transducer and activator of transcription-3 molecules is a reliable biomarker to quantify the photoreaction induced by PDT. Randomized studies are required to test the prognosis value of this biomarker, obtain approval for the new photosensitizers, and test the potential efficacy of interstitial and intraoperative PDT in the treatment of patients with non-small cell lung cancer.
Collapse
|
47
|
Boosting Tumor-Specific Immunity Using PDT. Cancers (Basel) 2016; 8:cancers8100091. [PMID: 27782066 PMCID: PMC5082381 DOI: 10.3390/cancers8100091] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/04/2016] [Indexed: 01/06/2023] Open
Abstract
Photodynamic therapy (PDT) is a cancer treatment with a long-standing history. It employs the application of nontoxic components, namely a light-sensitive photosensitizer and visible light, to generate reactive oxygen species (ROS). These ROS lead to tumor cell destruction, which is accompanied by the induction of an acute inflammatory response. This inflammatory process sends a danger signal to the innate immune system, which results in activation of specific cell types and release of additional inflammatory mediators. Activation of the innate immune response is necessary for subsequent induction of the adaptive arm of the immune system. This includes the priming of tumor-specific cytotoxic T lymphocytes (CTL) that have the capability to directly recognize and kill cells which display an altered self. The past decades have brought increasing appreciation for the importance of the generation of an adaptive immune response for long-term tumor control and induction of immune memory to combat recurrent disease. This has led to considerable effort to elucidate the immune effects PDT treatment elicits. In this review we deal with the progress which has been made during the past 20 years in uncovering the role of PDT in the induction of the tumor-specific immune response, with special emphasis on adaptive immunity.
Collapse
|
48
|
Yang J, Shen H, Jin H, Lou Q, Zhang X. Treatment of unresectable extrahepatic cholangiocarcinoma using hematoporphyrin photodynamic therapy: A prospective study. Photodiagnosis Photodyn Ther 2016; 16:110-118. [PMID: 27720942 DOI: 10.1016/j.pdpdt.2016.10.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 09/25/2016] [Accepted: 10/04/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND The available evidence of Photodynamic therapy (PDT) combined with stent placement treatment for unresectable extrahepatic cholangiocarcinoma (EHCC) is still insufficient. It also remains unclear whether PDT influences systemic inflammatory response. AIM To explore the clinical efficacy and safety of the combination treatment and the systemic inflammatory response in patients with EHCC. METHODS Patients with unresectable EHCC underwent either the combined treatment using Hematoporphyrin PDT and stent placement (PDT+stent group, n=12) or stent-only (stent group, n=27). The primary end-point was overall survival. Tumor necrosis factor (TNF)-α and interleukin (IL)-6 levels were measured. Quality of life was assessed using the Karnofsky performance scale (KPS) every 3 months. RESULTS Average survival time (13.8 vs. 9.6 months), and 6-month (91.7% vs. 74.1%), and 1-year (58.3% vs. 3.7%) survival rates of PDT+stent group were significantly increased compared with the stent group. KPS scores in the PDT+stent group were significantly improved. TNF-α and IL-6 levels were significantly increased in the PDT+stent group. CONCLUSION Hematoporphyrin-PDT combined with stent placement is an effective and safe treatment for EHCC. The treatment might promote systemic inflammatory response.
Collapse
Affiliation(s)
- Jianfeng Yang
- Department of Gastroenterology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang Province, China
| | - Hongzhang Shen
- Department of Gastroenterology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang Province, China
| | - Hangbin Jin
- Department of Gastroenterology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang Province, China
| | - Qifeng Lou
- Department of Gastroenterology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang Province, China
| | - Xiaofeng Zhang
- Department of Gastroenterology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
49
|
Malatesti N, Harej A, Kraljević Pavelić S, Lončarić M, Zorc H, Wittine K, Andjelkovic U, Josic D. Synthesis, characterisation and in vitro investigation of photodynamic activity of 5-(4-octadecanamidophenyl)-10,15,20-tris(N-methylpyridinium-3-yl)porphyrin trichloride on HeLa cells using low light fluence rate. Photodiagnosis Photodyn Ther 2016; 15:115-26. [PMID: 27431589 DOI: 10.1016/j.pdpdt.2016.07.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 07/10/2016] [Indexed: 01/09/2023]
Abstract
Photodynamic therapy (PDT) is a treatment that aims to kill cancer cells by reactive oxygen species, mainly singlet oxygen, produced through light activation of a photosensitiser (PS). Amongst photosensitisers that attracted the most attention in the last decade are cationic and amphiphilic molecules based on porphyrin, chlorin and phthalocyanine structures. Our aim was to join this search for more optimal balance of the lipophilic and hydrophilic moieties in a PS. A new amphiphilic porphyrin, 5-(4-octadecanamidophenyl)-10,15,20-tris(N-methylpyridinium-3-yl)porphyrin trichloride (5) was synthesised and characterised by (1)H NMR, UV-vis and fluorescence spectroscopy, and by MALDI-TOF/TOF spectrometry. In vitro photodynamic activity of 5 was evaluated on HeLa cell lines and compared to the activity of the hydrophilic 5-(4-acetamidophenyl)-10,15,20-tris(N-methylpyridinium-3-yl)porphyrin trichloride (7). Low fluence rate (2mWcm(-2)) of red light (643nm) was used for the activation, and both porphyrins showed a drug dose-response as well as a light dose-response relationship, but the amphiphilic porphyrin was presented with significantly lower IC50 values. The obtained IC50 values for 5 were 1.4μM at 15min irradiation time and 0.7μM when the time of irradiation was 30min, while for 7 these values were 37 and 6 times higher, respectively. These results confirm the importance of the lipophilic component in a PS and show a potential for 5 to be used as a PS in PDT applications.
Collapse
Affiliation(s)
- Nela Malatesti
- Department of Biotechnology, Centre for high-throughput technologies, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia.
| | - Anja Harej
- Department of Biotechnology, Centre for high-throughput technologies, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia.
| | - Sandra Kraljević Pavelić
- Department of Biotechnology, Centre for high-throughput technologies, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia.
| | - Martin Lončarić
- Laboratory for Photonics and Quantum Optics, Division of Experimental Physics, Ruđer Bošković Institute, Bijenička cesta 54, 10002 Zagreb, Croatia.
| | - Hrvoje Zorc
- Laboratory of Optics and Optical Thin Films, Division of Materials Physics, Ruđer Bošković Institute, Bijenička cesta 54, 10002 Zagreb, Croatia.
| | - Karlo Wittine
- Department of Biotechnology, Centre for high-throughput technologies, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia.
| | - Uros Andjelkovic
- Department of Biotechnology, Centre for high-throughput technologies, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia.
| | - Djuro Josic
- Department of Biotechnology, Centre for high-throughput technologies, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia.
| |
Collapse
|
50
|
White B, Rossi V, Baugher PJ. Aminolevulinic Acid-Mediated Photodynamic Therapy Causes Cell Death in MG-63 Human Osteosarcoma Cells. Photomed Laser Surg 2016; 34:400-5. [PMID: 27500317 DOI: 10.1089/pho.2016.4091] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE The aim of this study was to test the efficacy of aminolevulinic acid-mediated photodynamic therapy (PDT) against the human osteosarcoma cell line MG-63. BACKGROUND DATA Osteosarcoma is the most common type of primary malignant bone tumor diagnosed in the United States among adolescents and children. Treatments for osteosarcoma often result in diminished limb use or amputation. Because ALA-mediated PDT exhibits dual specificity in the context of tumor killing, this therapy could represent a less invasive, but effective, treatment for this disease. MATERIALS AND METHODS To assess ALA dark toxicity in MG-63 cells, cells were incubated with varying concentrations of ALA, and cell viability was determined by crystal violet assay. Protoporphyrin IX (PpIX) accumulation was assessed subsequent to ALA incubation at various concentrations using spectrofluorometry. Cell death subsequent to ALA-PDT was determined by illuminating cells at a wavelength of 635 nm at various light intensities subsequent to ALA incubation. Cell viability was assessed using the MTT assay. RESULTS ALA dark toxicity was observed only at the highest concentrations of 2, 5, and 10 mM. Maximal PpIX concentration was observed at 0.5 and 1 mM ALA, subsequent to a 24-h incubation. Maximal cell death with minimal light toxicity was observed at 0.5 and 1 mM ALA after illumination with 0.6 and 3 J/cm(2) light. CONCLUSIONS Collectively, our data indicate that ALA-PDT can result in the death of MG-64 human osteosarcoma cells in vitro.
Collapse
Affiliation(s)
- Bradley White
- 1 Department of Biology, School of Natural Sciences, College of Arts and Sciences, Pacific University , Forest Grove, Oregon
| | - Vince Rossi
- 2 Department of Physics, School of Natural Sciences, College of Arts and Sciences, Pacific University , Forest Grove, Oregon
| | - Paige J Baugher
- 1 Department of Biology, School of Natural Sciences, College of Arts and Sciences, Pacific University , Forest Grove, Oregon
| |
Collapse
|