1
|
Nahm WJ, Sakunchotpanit G, Nambudiri VE. Abscopal Effects and Immunomodulation in Skin Cancer Therapy. Am J Clin Dermatol 2025:10.1007/s40257-025-00943-x. [PMID: 40180765 DOI: 10.1007/s40257-025-00943-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2025] [Indexed: 04/05/2025]
Abstract
Radiation therapy (RT) is a crucial modality in cancer treatment, functioning through direct DNA damage and immune stimulation. However, RT's effects extend beyond targeted cells, influencing neighboring cells through the bystander effect (ByE) and distant sites via the abscopal effect (AbE). The AbE, first described by Mole in 1953, encompasses biological reactions at sites distant from the irradiation field. While RT can enhance antitumor immune responses, it may also contribute to an immunosuppressive microenvironment. To address this limitation, combining RT with immune checkpoint inhibitors (ICIs) has gained renewed interest, aiming to amplify antitumor immune responses. Evidence of AbEs has been observed in various metastatic or advanced cutaneous cancers, including melanoma, basal cell carcinoma, cutaneous lymphoma, Merkel cell carcinoma, and cutaneous squamous cell carcinoma. Clinical studies suggest combining RT with ICIs targeting CTLA-4 and PD-1/PD-L1 may enhance AbE incidence in these cancers. This review primarily explores the current understanding of AbEs in skin cancers, briefly acknowledging the ByE focusing on combining RT with immunomodulation. It focuses on proposed mechanisms, preclinical and clinical evidence, challenges in clinical translation, and future directions for harnessing AbEs in managing advanced skin malignancies. Alternative modalities for inducing abscopal-like responses are also explored. While promising, challenges remain in consistently reproducing AbEs in clinical practice, necessitating further research to optimize treatment combinations, timing, and patient selection.
Collapse
Affiliation(s)
- William J Nahm
- New York University Grossman School of Medicine, New York, NY, USA.
- Department of Dermatology, Brigham and Women's Hospital, 117 Western Avenue, Boston, MA, 02163, USA.
| | - Goranit Sakunchotpanit
- Department of Dermatology, Brigham and Women's Hospital, 117 Western Avenue, Boston, MA, 02163, USA
- Tufts University School of Medicine, Boston, MA, USA
| | - Vinod E Nambudiri
- Department of Dermatology, Brigham and Women's Hospital, 117 Western Avenue, Boston, MA, 02163, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
2
|
Jafri Z, Li Y, Zhang J, O’Meara CH, Khachigian LM. Jun, an Oncological Foe or Friend? Int J Mol Sci 2025; 26:555. [PMID: 39859271 PMCID: PMC11766113 DOI: 10.3390/ijms26020555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 01/27/2025] Open
Abstract
Jun/JUN is a basic leucine zipper (bZIP) protein and a prototypic member of the activator protein-1 (AP-1) family of transcription factors that can act as homo- or heterodimers, interact with DNA elements and co-factors, and regulate gene transcription. Jun is expressed by both immune and inflammatory cells. Jun is traditionally seen as an oncoprotein that regulates processes involved in transformation and oncogenesis in human tumours. This article examines the traditional view that Jun plays a permissive role in cancer development and progression, whilst exploring emerging evidence supporting Jun's potential to prevent immune cell exhaustion and promote anti-tumour efficacy.
Collapse
Affiliation(s)
- Zuhayr Jafri
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Yue Li
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jingwen Zhang
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Connor H. O’Meara
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Division of Head & Neck Oncology and Microvascular Reconstruction, Department of Otolaryngology, Head & Neck Surgery, University of Virginia Health Services, Charlottesville, VA 22903, USA
- Department of Otolaryngology, Head & Neck Surgery, Australian National University, Acton, ACT 0200, Australia
| | - Levon M. Khachigian
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
3
|
Wu X, Li Y, Wen M, Xie Y, Zeng K, Liu YN, Chen W, Zhao Y. Nanocatalysts for modulating antitumor immunity: fabrication, mechanisms and applications. Chem Soc Rev 2024; 53:2643-2692. [PMID: 38314836 DOI: 10.1039/d3cs00673e] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Immunotherapy harnesses the inherent immune system in the body to generate systemic antitumor immunity, offering a promising modality for defending against cancer. However, tumor immunosuppression and evasion seriously restrict the immune response rates in clinical settings. Catalytic nanomedicines can transform tumoral substances/metabolites into therapeutic products in situ, offering unique advantages in antitumor immunotherapy. Through catalytic reactions, both tumor eradication and immune regulation can be simultaneously achieved, favoring the development of systemic antitumor immunity. In recent years, with advancements in catalytic chemistry and nanotechnology, catalytic nanomedicines based on nanozymes, photocatalysts, sonocatalysts, Fenton catalysts, electrocatalysts, piezocatalysts, thermocatalysts and radiocatalysts have been rapidly developed with vast applications in cancer immunotherapy. This review provides an introduction to the fabrication of catalytic nanomedicines with an emphasis on their structures and engineering strategies. Furthermore, the catalytic substrates and state-of-the-art applications of nanocatalysts in cancer immunotherapy have also been outlined and discussed. The relationships between nanostructures and immune regulating performance of catalytic nanomedicines are highlighted to provide a deep understanding of their working mechanisms in the tumor microenvironment. Finally, the challenges and development trends are revealed, aiming to provide new insights for the future development of nanocatalysts in catalytic immunotherapy.
Collapse
Affiliation(s)
- Xianbo Wu
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - Yuqing Li
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - Mei Wen
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - Yongting Xie
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - Ke Zeng
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - You-Nian Liu
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - Wansong Chen
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China.
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore.
| |
Collapse
|
4
|
Liu H, Li Y, Du S, Wang C, Li Y, Cao R, Shi W, Liu S, He J. Studies on the Effect of Lipofectamine and Cell-Penetrating Peptide on the Properties of 10-23 DNAzyme. Molecules 2023; 28:molecules28093942. [PMID: 37175352 PMCID: PMC10179765 DOI: 10.3390/molecules28093942] [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: 03/14/2023] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Cationic polymeric materials and cell-penetrating peptides (CPPs) were often used as the delivery vectors in the evaluation of nucleic acid therapeutics. 10-23 DNAzyme is a kind of potential antisense therapeutics by catalytic cleavage of the disease-related RNAs. Here, lipofectamine 2000 and Tat peptide were evaluated for their effect on the catalytic activity of 10-23 DNAzyme, with the observed rate constant, thermal stability, CD spectra, and PAGE analysis, with a duplex DNA mimicking DNAzyme-substrate as a control. It was shown that the cationic carriers had a negative effect on the catalytic performance of the 10-23 DNAzyme. Significantly, the destabilizing effect of the cationic carriers on the duplex formation was noteworthy, as a duplex formation is an essential prerequisite in the silencing mechanisms of antisense and RNAi.
Collapse
Affiliation(s)
- Huanhuan Liu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
- State Key Laboratory of Toxicology and Medical Countermeasurements, Beijing Institute of Pharmacology and Toxicology, Taiping 27, Beijing 100850, China
| | - Yang Li
- State Key Laboratory of Toxicology and Medical Countermeasurements, Beijing Institute of Pharmacology and Toxicology, Taiping 27, Beijing 100850, China
| | - Shanshan Du
- State Key Laboratory of Toxicology and Medical Countermeasurements, Beijing Institute of Pharmacology and Toxicology, Taiping 27, Beijing 100850, China
| | - Chenhong Wang
- State Key Laboratory of Toxicology and Medical Countermeasurements, Beijing Institute of Pharmacology and Toxicology, Taiping 27, Beijing 100850, China
| | - Yuexiang Li
- State Key Laboratory of Toxicology and Medical Countermeasurements, Beijing Institute of Pharmacology and Toxicology, Taiping 27, Beijing 100850, China
| | - Ruiyuan Cao
- State Key Laboratory of Toxicology and Medical Countermeasurements, Beijing Institute of Pharmacology and Toxicology, Taiping 27, Beijing 100850, China
| | - Weiguo Shi
- State Key Laboratory of Toxicology and Medical Countermeasurements, Beijing Institute of Pharmacology and Toxicology, Taiping 27, Beijing 100850, China
| | - Shihui Liu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Junlin He
- State Key Laboratory of Toxicology and Medical Countermeasurements, Beijing Institute of Pharmacology and Toxicology, Taiping 27, Beijing 100850, China
| |
Collapse
|
5
|
Rinaldi-Neto F, Ribeiro AB, Ferreira NH, Squarisi IS, Oliveira KM, Orenha RP, Parreira RLT, Batista AA, Tavares DC. Anti-melanoma effect of ruthenium(II)-diphosphine complexes containing naphthoquinone ligand. J Inorg Biochem 2021; 222:111497. [PMID: 34090039 DOI: 10.1016/j.jinorgbio.2021.111497] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 12/18/2022]
Abstract
The use of natural products as potential ligands has been explored as a strategy in the development of metal-based chemotherapy. Since ruthenium complexes are promising alternatives to traditional antitumor agents, this study evaluated the anti-melanoma potential of two ruthenium(II) complexes containing the naphthoquinone ligands lapachol (lap), [Ru(lap)(dppm)2]PF6, and lawsone (law), [Ru(law)(dppm)2]PF6, in addition to the bis(diphenylphosphino)methane (dppm) ligand, referred to as complexes (1) and (2), respectively, using a syngeneic murine melanoma model. Activation of the apoptotic pathway by the treatments was assessed by immunohistochemistry in tumor tissue. Additionally, toxicity of the treatments was evaluated by variation in body and organ weight, quantification of biochemical indicators of renal damage, and genotoxicity in bone marrow and hepatocytes. First, the antiproliferative activity of (1) and (2) was observed in B16F10 cells, with IC50 values of 2.78 and 1.68 μM, respectively. The results obtained in mice showed that, unlike complex (1), (2) possesses significant anti-melanoma activity demonstrated by a reduction in tumor volume and mass (88.42%), as well as in mitosis frequency (83.86%). Additionally, complex (2) increased the levels of cleaved caspase-3, inducing tumor cell apoptosis. When compared to the metallodrug cisplatin, complex (2) exhibited similar anti-melanoma activity and lower toxicity considering all parameters evaluated. In silico studies demonstrated no difference in the binding energy of the naphthoquinone complex between complexes (1) and (2). However, the complex containing the lawsone ligand has a lower molar volume, which may be important for interactions with minor DNA grooves. The present results demonstrate the antitumor efficiency of complex (2) and a significantly lower systemic toxicity compared to cisplatin.
Collapse
Affiliation(s)
- Francisco Rinaldi-Neto
- Universidade de Franca, Avenida Dr. Armando Salles Oliveira, 201 - Parque Universitário, Franca, São Paulo 14404-600, Brazil
| | - Arthur Barcelos Ribeiro
- Universidade de Franca, Avenida Dr. Armando Salles Oliveira, 201 - Parque Universitário, Franca, São Paulo 14404-600, Brazil
| | - Natália Helen Ferreira
- Universidade de Franca, Avenida Dr. Armando Salles Oliveira, 201 - Parque Universitário, Franca, São Paulo 14404-600, Brazil
| | - Iara Silva Squarisi
- Universidade de Franca, Avenida Dr. Armando Salles Oliveira, 201 - Parque Universitário, Franca, São Paulo 14404-600, Brazil
| | - Kátia Mara Oliveira
- Universidade Federal de São Carlos, Departamento de Química, Rodovia Washington Luis s/n Km 235, São Carlos, São Paulo 13565-905, Brazil
| | - Renato Pereira Orenha
- Universidade de Franca, Avenida Dr. Armando Salles Oliveira, 201 - Parque Universitário, Franca, São Paulo 14404-600, Brazil
| | - Renato Luís Tame Parreira
- Universidade de Franca, Avenida Dr. Armando Salles Oliveira, 201 - Parque Universitário, Franca, São Paulo 14404-600, Brazil
| | - Alzir Azevedo Batista
- Universidade Federal de São Carlos, Departamento de Química, Rodovia Washington Luis s/n Km 235, São Carlos, São Paulo 13565-905, Brazil
| | - Denise Crispim Tavares
- Universidade de Franca, Avenida Dr. Armando Salles Oliveira, 201 - Parque Universitário, Franca, São Paulo 14404-600, Brazil.
| |
Collapse
|
6
|
Huo W, Li X, Wang B, Zhang H, Zhang J, Yang X, Jin Y. Recent advances of DNAzyme-based nanotherapeutic platform in cancer gene therapy. BIOPHYSICS REPORTS 2020. [DOI: 10.1007/s41048-020-00123-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
AbstractDeoxyribozyme (or denoted as DNAzyme), which is produced by in vitro screening technology, has gained extensive research interest in the field of biomedicine due to its high catalytic activity and structure identification. This review introduces the structural characteristics of RNA-cleaving DNAzyme and its application potential in cancer gene therapy, which plays a significant role in cancer-related gene inactivation by specifically cleaving target mRNA and inhibiting the expression of the corresponding protein. However, the low delivery efficiency and cellular uptake hindered the widespread usage of DNAzyme in gene therapy of cancers. Emerging nanotechnology holds great promise for DNAzyme to overcome these obstacles. This review mainly focuses on DNAzyme-based nanotherapeutic platforms in gene therapy of cancers, including oncogene antagonism therapy, treatment resistance gene therapy, immunogene therapy, and antiangiogenesis gene therapy. We also revealed the potential of DNAzyme-based nanotherapeutic platforms as emerging cancer therapy approaches and their security issues.
Collapse
|
7
|
Castro KADF, Costa LD, Guieu S, Biazzotto JC, da Neves MGPMS, Faustino MAF, da Silva RS, Tomé AC. Photodynamic treatment of melanoma cells using aza-dipyrromethenes as photosensitizers. Photochem Photobiol Sci 2020; 19:885-891. [PMID: 32662457 DOI: 10.1039/d0pp00114g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In this study, we report for the first time the use of four aza-dipyrromethenes (ADPMs) as photosensitizers for cancer PDT. The synthesis and characterization of the ADPMs and their photodynamic action against B16F10 melanoma cells were assessed. ADPM 2 is the best singlet oxygen generator and the most phototoxic (at 2.5 μM) towards B16F10 cells.
Collapse
Affiliation(s)
- Kelly A D F Castro
- Department of Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, SP, Brazil.
| | - Letícia D Costa
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Portugal
| | - Samuel Guieu
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Portugal.,CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Portugal
| | - Juliana C Biazzotto
- Department of Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, SP, Brazil
| | | | | | - Roberto S da Silva
- Department of Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, SP, Brazil
| | - Augusto C Tomé
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Portugal
| |
Collapse
|
8
|
Kappelmann-Fenzl M, Kuphal S, Krupar R, Schadendorf D, Umansky V, Vardimon L, Hellerbrand C, Bosserhoff AK. Complex Formation with Monomeric α-Tubulin and Importin 13 Fosters c-Jun Protein Stability and Is Required for c-Jun's Nuclear Translocation and Activity. Cancers (Basel) 2019; 11:cancers11111806. [PMID: 31744174 PMCID: PMC6895814 DOI: 10.3390/cancers11111806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 12/25/2022] Open
Abstract
Microtubules are highly dynamic structures, which consist of α- and β-tubulin heterodimers. They are essential for a number of cellular processes, including intracellular trafficking and mitosis. Tubulin-binding chemotherapeutics are used to treat different types of tumors, including malignant melanoma. The transcription factor c-Jun is a central driver of melanoma development and progression. Here, we identify the microtubule network as a main regulator of c-Jun activity. Monomeric α-tubulin fosters c-Jun protein stability by protein-protein interaction. In addition, this complex formation is necessary for c-Jun's nuclear localization sequence binding to importin 13, and consequent nuclear import and activity of c-Jun. A reduction in monomeric α-tubulin levels by treatment with the chemotherapeutic paclitaxel resulted in a decline in the nuclear accumulation of c-Jun in melanoma cells in an experimental murine model and in patients' tissues. These findings add important knowledge to the mechanism of the action of microtubule-targeting drugs and indicate the newly discovered regulation of c-Jun by the microtubule cytoskeleton as a novel therapeutic target for melanoma and potentially also other types of cancer.
Collapse
Affiliation(s)
- Melanie Kappelmann-Fenzl
- Institute of Biochemistry (Emil-Fischer Center), Friedrich-Alexander University, Erlangen-Nürnberg, 91054 Erlangen, Germany (S.K.); (C.H.)
- Faculty of Applied Health Care Sciences, University of Applied Science Deggendorf, 94469 Deggendorf, Germany
| | - Silke Kuphal
- Institute of Biochemistry (Emil-Fischer Center), Friedrich-Alexander University, Erlangen-Nürnberg, 91054 Erlangen, Germany (S.K.); (C.H.)
| | - Rosemarie Krupar
- Pathology of the University Medical Center Schleswig-Holstein, Campus Lübeck and Research Center Borstel, Leibniz Center for Medicine and Biosciences, 23566 Lübeck, Germany;
| | - Dirk Schadendorf
- Department of Dermatology, University Duisburg-Essen, 45355 Essen, Germany;
| | - Viktor Umansky
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, 69117 Heidelberg, Germany;
| | - Lily Vardimon
- Department of Biochemistry and Molecular Biology, Tel Aviv University, 69978 Tel Aviv, Israel;
| | - Claus Hellerbrand
- Institute of Biochemistry (Emil-Fischer Center), Friedrich-Alexander University, Erlangen-Nürnberg, 91054 Erlangen, Germany (S.K.); (C.H.)
- Comprehensive Cancer Center (CCC) Erlangen-EMN, 91054 Erlangen, Germany
| | - Anja-Katrin Bosserhoff
- Institute of Biochemistry (Emil-Fischer Center), Friedrich-Alexander University, Erlangen-Nürnberg, 91054 Erlangen, Germany (S.K.); (C.H.)
- Comprehensive Cancer Center (CCC) Erlangen-EMN, 91054 Erlangen, Germany
- Correspondence: ; Tel.: +49-9131-8524191
| |
Collapse
|
9
|
Xiao L, Gu C, Xiang Y. Orthogonal Activation of RNA‐Cleaving DNAzymes in Live Cells by Reactive Oxygen Species. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201908105] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Lu Xiao
- Department of Chemistry Beijing Key Laboratory for Microanalytical Methods and Instrumentation Key Laboratory of Bioorganic Phosphorus, Chemistry and Chemical Biology (Ministry of Education) Tsinghua University Beijing 100084 China
| | - Chunmei Gu
- Department of Chemistry Beijing Key Laboratory for Microanalytical Methods and Instrumentation Key Laboratory of Bioorganic Phosphorus, Chemistry and Chemical Biology (Ministry of Education) Tsinghua University Beijing 100084 China
| | - Yu Xiang
- Department of Chemistry Beijing Key Laboratory for Microanalytical Methods and Instrumentation Key Laboratory of Bioorganic Phosphorus, Chemistry and Chemical Biology (Ministry of Education) Tsinghua University Beijing 100084 China
| |
Collapse
|
10
|
Xiao L, Gu C, Xiang Y. Orthogonal Activation of RNA-Cleaving DNAzymes in Live Cells by Reactive Oxygen Species. Angew Chem Int Ed Engl 2019; 58:14167-14172. [PMID: 31314942 DOI: 10.1002/anie.201908105] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Indexed: 02/05/2023]
Abstract
RNA-cleaving DNAzymes are useful tools for intracellular metal-ion sensing and gene regulation. Incorporating stimuli-responsive modifications into these DNAzymes enables their activities to be spatiotemporally and chemically controlled for more precise applications. Despite the successful development of many caged DNAzymes for light-induced activation, DNAzymes that can be intracellularly activated by chemical inputs of biological importance, such as reactive oxygen species (ROS), are still scarce. ROS like hydrogen peroxide (H2 O2 ) and hypochlorite (HClO) are critical mediators of oxidative stress-related cell signaling and dysregulation including activation of immune system as well as progression of diseases and aging. Herein, we report ROS-activable DNAzymes by introducing phenylboronate and phosphorothioate modifications to the Zn2+ -dependent 8-17 DNAzyme. These ROS-activable DNAzymes were orthogonally activated by H2 O2 and HClO inside live human and mouse cells.
Collapse
Affiliation(s)
- Lu Xiao
- Department of Chemistry, Beijing Key Laboratory for Microanalytical Methods and Instrumentation, Key Laboratory of Bioorganic Phosphorus, Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, China
| | - Chunmei Gu
- Department of Chemistry, Beijing Key Laboratory for Microanalytical Methods and Instrumentation, Key Laboratory of Bioorganic Phosphorus, Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, China
| | - Yu Xiang
- Department of Chemistry, Beijing Key Laboratory for Microanalytical Methods and Instrumentation, Key Laboratory of Bioorganic Phosphorus, Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, China
| |
Collapse
|
11
|
Verteporfin-loaded mesoporous silica nanoparticles inhibit mouse melanoma proliferation in vitro and in vivo. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 197:111533. [PMID: 31254952 DOI: 10.1016/j.jphotobiol.2019.111533] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/07/2019] [Accepted: 06/12/2019] [Indexed: 12/22/2022]
Abstract
Melanoma is one of the most lethal tumors among the skin cancers, arising from complex genetic mutations in melanocyte. Melanoma microenvironment is very heterogeneous, showing complex vascular networks and immunogenicity, as well as induced acquired resistance to treatments by upregulation of multidrug resistance (MDR) mechanisms. Different studies have showed that Photodynamic Therapy (PDT) could be considered a new potential approach for melanoma treatment. PDT combines a light with a specific wavelength and a photosensitizer: when these two elements interact reactive oxygen species (ROS) are generated leading to tumor cell destruction. In this study verteporfin (Ver), a second-generation photosensitizer, has been conjugated with mesoporous silica nanoparticles (MSNs): the resulting Ver-MSNs are an efficient nanoplatforms used to enhance cargo capacity and cellular uptake. Our in vitro and in vivo studies investigated whether Ver-MSNs were able to reduce or inhibit melanoma growth. In vitro experiments performed using B16F10 mouse melanoma cells showed that Ver-MSNs stimulated by red light (693 nm) significantly decreased in vitro cells proliferation in a range of concentration between 0.1 μg/ml to 10 μg/ml. When Ver-MSNs (5 μg/ml in glycerol) were topically administrated to melanoma tumor mass developed in mice and stimulated by red light for four times in 16 days, they were able to reduce the tumor mass of 50.2 ± 6,6% compared to the untreated (only glycerol) mice. In the light of this information, PDT performed using Ver-MSNs could be considered a new promising and potential approach to treat melanoma.
Collapse
|
12
|
Khachigian LM. Deoxyribozymes as Catalytic Nanotherapeutic Agents. Cancer Res 2019; 79:879-888. [DOI: 10.1158/0008-5472.can-18-2474] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/24/2018] [Accepted: 12/14/2018] [Indexed: 11/16/2022]
|
13
|
RNA-Cleaving DNAzymes: Old Catalysts with New Tricks for Intracellular and In Vivo Applications. Catalysts 2018. [DOI: 10.3390/catal8110550] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
DNAzymes are catalytically active DNA molecules that are normally isolated through in vitro selection methods, among which RNA-cleaving DNAzymes that catalyze the cleavage of a single RNA linkage embedded within a DNA strand are the most studied group of this DNA enzyme family. Recent advances in DNA nanotechnology and engineering have generated many RNA-cleaving DNAzymes with unique recognition and catalytic properties. Over the past decade, numerous RNA-cleaving, DNAzymes-based functional probes have been introduced into many research areas, such as in vitro diagnostics, intracellular imaging, and in vivo therapeutics. This review focus on the fundamental insight into RNA-Cleaving DNAzymes and technical tricks for their intracellular and in vivo applications, highlighting the recent progress in the clinical trial of RNA-Cleaving DNAzymes with selected examples. The challenges and opportunities for the future translation of RNA-cleaving DNAzymes for biomedicine are also discussed.
Collapse
|