1
|
Repetowski P, Warszyńska M, Dąbrowski JM. NIR-activated multifunctional agents for the combined application in cancer imaging and therapy. Adv Colloid Interface Sci 2025; 336:103356. [PMID: 39612723 DOI: 10.1016/j.cis.2024.103356] [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: 08/16/2024] [Accepted: 11/17/2024] [Indexed: 12/01/2024]
Abstract
Anticancer therapies that combine both diagnostic and therapeutic capabilities hold significant promise for enhancing treatment efficacy and patient outcomes. Among these, agents responsive to near-infrared (NIR) photons are of particular interest due to their negligible toxicity and multifunctionality. These compounds are not only effective in photodynamic therapy (PDT), but also serve as contrast agents in various imaging modalities, including fluorescence and photoacoustic imaging. In this review, we explore the photophysical and photochemical properties of NIR-activated porphyrin, cyanine, and phthalocyanines derivatives as well as aggregation-induced emission compounds, highlighting their application in synergistic detection, diagnosis, and therapy. Special attention is given to the design and optimization of these agents to achieve high photostability, efficient NIR absorption, and significant yields of fluorescence, heat, or reactive oxygen species (ROS) generation depending on the application. Additionally, we discuss the incorporation of these compounds into nanocarriers to enhance their solubility, stability, and target specificity. Such nanoparticle-based systems exhibit improved pharmacokinetics and pharmacodynamics, facilitating more effective tumor targeting and broadening the application range to photoacoustic imaging and photothermal therapy. Furthermore, we summarize the application of these NIR-responsive agents in multimodal imaging techniques, which combine the advantages of fluorescence and photoacoustic imaging to provide comprehensive diagnostic information. Finally, we address the current challenges and limitations of photodiagnosis and phototherapy and highlight some critical barriers to their clinical implementation. These include issues related to their phototoxicity, limited tissue penetration, and potential off-target effects. The review concludes by highlighting future research directions aimed at overcoming these obstacles, with a focus on the development of next-generation agents and platforms that offer enhanced therapeutic efficacy and imaging capabilities in the field of cancer treatment.
Collapse
Affiliation(s)
- Paweł Repetowski
- Faculty of Chemistry, Jagiellonian University, 30-387 Kraków, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-348 Kraków, Poland
| | - Marta Warszyńska
- Faculty of Chemistry, Jagiellonian University, 30-387 Kraków, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-348 Kraków, Poland
| | | |
Collapse
|
2
|
Liao Y, Lin X, He Z, Chen J, Tang S, Wang W, Chen W. Construction of nucleus-targeted photosensitizer and highly effective photodynamic immunotherapy for cancer. Bioorg Chem 2025; 154:108022. [PMID: 39642756 DOI: 10.1016/j.bioorg.2024.108022] [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: 10/14/2024] [Revised: 11/20/2024] [Accepted: 11/29/2024] [Indexed: 12/09/2024]
Abstract
Nucleus is the largest and most important organelle within eukaryotic cells, containing most of the cell's genetic material, DNA. It serves as the central hub for genetic regulation and metabolism, making it an ideal target for subcellular drug delivery. The development of nucleus-targeted photosensitizers allows for the rapid and effective destruction of critical components such as DNA within the nucleus. This achieves the goal of efficiently eliminating cancer cells. However, most organic molecules, including photosensitizers, cannot penetrate the nuclear membrane, making the design and synthesis of nucleus-targeted photosensitizers both significant and challenging. The authors have designed and synthesized a nucleus-targeted activatable photosensitive probe (CMT-I). In vitro spectral analyses demonstrate that CMT-I is specifically activated by ct-DNA, significantly enhancing fluorescence-a 49-fold increase is observed upon binding. Furthermore, under 590 nm light irradiation, CMT-I effectively generates 1O2. Molecular docking show that CMT-I selectively binds to DNA through hydrogen bonds and ᴨ-ᴨ conjugation. RNA sequencing experiments reveal that photodynamic therapy activates immunity within tumor cells, triggering an adaptive immune response. In vivo therapeutic experiments further verify the enhanced anti-tumor immunity of CMT-I, which is crucial for effectively eliminating immunologically cold tumors and highlights the potential of DNA-targeted photodynamic therapy in precise cancer treatment.
Collapse
Affiliation(s)
- Yacong Liao
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Xiaoping Lin
- College of Chemistry and Material Science, Hengyang Normal University, Hengyang 421001, PR China
| | - Zhenhu He
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Juan Chen
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Siping Tang
- College of Chemistry and Material Science, Hengyang Normal University, Hengyang 421001, PR China
| | - Wei Wang
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha 410008, PR China.
| | - Wen Chen
- College of Chemistry and Material Science, Hengyang Normal University, Hengyang 421001, PR China.
| |
Collapse
|
3
|
Bel'ko N, Mal'tanova A, Bahdanava A, Lugovski A, Fatykhava S, Shabunya P, Smaliakou A, Poznyak S, Kulahava T, Samtsov M. A near-infrared superoxide generator based on a biocompatible indene-bearing heptamethine cyanine dye. J Mater Chem B 2024; 12:11202-11209. [PMID: 39364565 DOI: 10.1039/d4tb01663g] [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: 10/05/2024]
Abstract
One of the most significant limitations of photodynamic therapy is its reduced efficacy in hypoxic microenvironments, which are typical of the majority of tumors. This work demonstrates that indolenine heptamethine cyanines with different substituents in the polymethine chain and at the terminal heterocycles are effective superoxide generators that can be activated in the near-infrared range. The introduction of an indene moiety into the polymethine chain results in a significant enhancement in photostability compared to dyes with a cyclohexene moiety or an unsubstituted polymethine chain. A hydrophilic indene-bearing heptamethine cyanine dye is shown to be efficiently internalized by Vero E6 cells and to give bright intracellular fluorescence in the 700-850 nm range. Furthermore, the dye generates superoxide anion radicals and induces severe oxidative stress in cells upon activation in the near-infrared range (∼750 nm), ultimately resulting in cell death. The capacity of heptamethine cyanines to generate a superoxide anion radical may prove advantageous for enhancing the efficacy of photodynamic therapy under hypoxic conditions.
Collapse
Affiliation(s)
- Nikita Bel'ko
- A.N. Sevchenko Institute of Applied Physical Problems, Belarusian State University, Kurchatova str. 7, Minsk 220045, Belarus.
| | - Anna Mal'tanova
- Research Institute for Physical Chemical Problems, Belarusian State University, Leningradskaya str. 14, Minsk 220006, Belarus
| | - Anastasiya Bahdanava
- Institute for Nuclear Problems, Belarusian State University, Bobruiskaya str. 11, Minsk 220006, Belarus
| | - Anatol Lugovski
- A.N. Sevchenko Institute of Applied Physical Problems, Belarusian State University, Kurchatova str. 7, Minsk 220045, Belarus.
| | - Sviatlana Fatykhava
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Akademika Kuprevicha str. 5-2, Minsk 220141, Belarus
| | - Polina Shabunya
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Akademika Kuprevicha str. 5-2, Minsk 220141, Belarus
| | - Adam Smaliakou
- Department of Physics, Belarusian State University, Bobruiskaya str. 5, Minsk 220006, Belarus
| | - Sergey Poznyak
- Research Institute for Physical Chemical Problems, Belarusian State University, Leningradskaya str. 14, Minsk 220006, Belarus
| | - Tatsiana Kulahava
- Institute for Nuclear Problems, Belarusian State University, Bobruiskaya str. 11, Minsk 220006, Belarus
| | - Michael Samtsov
- A.N. Sevchenko Institute of Applied Physical Problems, Belarusian State University, Kurchatova str. 7, Minsk 220045, Belarus.
| |
Collapse
|
4
|
Nasr Esfahani F, Karimi S, Jalilian Z, Alavi M, Aziz B, Alhagh Charkhat Gorgich E, Mozafari MR, Taghavi E, Aminnezhad S, Ataei S. Functionalized and Theranostic Lipidic and Tocosomal Drug Delivery Systems: Potentials and Limitations in Cancer Photodynamic Therapy. Adv Pharm Bull 2024; 14:524-536. [PMID: 39494248 PMCID: PMC11530887 DOI: 10.34172/apb.2024.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 02/09/2024] [Accepted: 03/03/2024] [Indexed: 11/05/2024] Open
Abstract
Photodynamic therapy (PDT) is a multidisciplinary area, which involves photophysics and photochemical sciences and plays an important role in cancer diagnosis and treatment. PDT involves a photo-activable drug called photosensitizer (PS), a specific wavelength of light and cellular compounds to produce toxic oxygen species in a much-localized way to destroy malignant tumors. Despite the various benefits of PDT, some PS-related limitations hinder its use as an ideal treatment option for cancer. To address these limitations (e.g., poor bioavailability, weak permeability, hydrophobicity, and aggregation), lipid-based and vesicular drug delivery systems have been employed. These carrier systems possess the ability to enhance the bioavailability, permeability, and solubility of the drug. Furthermore, they tend to load hydrophobic and lipophilic compounds and can be employed for an efficient and targeted drug delivery. The purpose of this review is to highlight the precise idea of PDT, the limitations of PDT related to PS, and the application of lipidic and tocosomal carriers in PDT for the treatment of various types of cancers. Liposomes, nanoliposomes, solid lipid nanoparticles, vesicular phospholipid gels, exosomes, transferosomes, and tocosomes are presented as commonly-employed vesicular drug carriers. Moreover, the amalgamation of cell-based drug delivery systems (CBDDS) with PDT holds considerable potential as an encouraging avenue in cancer treatment, especially in the context of immunotherapy.
Collapse
Affiliation(s)
- Fahime Nasr Esfahani
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| | - Sahand Karimi
- Department of Biological Science, Faculty of Science, University of Kurdistan, Sanandaj, Kurdistan 6617715175, Iran
| | - Zahra Jalilian
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| | - Mehran Alavi
- Department of Biological Science, Faculty of Science, University of Kurdistan, Sanandaj, Kurdistan 6617715175, Iran
| | - Bushra Aziz
- Department of Physics, Women University of Azad Jammu & Kashmir, Bagh 12500, Azad Kashmir, Pakistan
| | - Enam Alhagh Charkhat Gorgich
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - M. R. Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| | - Elham Taghavi
- Faculty of Fisheries and Food Science, Universiti Malaysia Terengganu (UMT), 21030 Kuala Nerus, Terengganu, Malaysia
| | - Sargol Aminnezhad
- Department of Microbiology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Sara Ataei
- Department of Clinical Pharmacy (Pharmacotherapy), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Maimaitijiang A, He D, Li D, Li W, Su Z, Fan Z, Li J. Progress in Research of Nanotherapeutics for Overcoming Multidrug Resistance in Cancer. Int J Mol Sci 2024; 25:9973. [PMID: 39337463 PMCID: PMC11432649 DOI: 10.3390/ijms25189973] [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: 07/28/2024] [Revised: 09/12/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
Chemotherapy has been widely applied in oncotherapy. However, the development of multidrug resistance (MDR) has diminished the effectiveness of anticancer drugs against tumor cells. Such resistance often results in tumor recurrence, metastasis, and patient death. Fortunately, nanoparticle-based drug delivery systems provide a promising strategy by codelivery of multiple drugs and MDR reversal agents and the skillful, flexible, smart modification of drug targets. Such systems have demonstrated the ability to bypass the ABC transporter biological efflux mechanisms due to drug resistance. Hence, how to deliver drugs and exert potential antitumor effects have been successfully explored, applied, and developed. Furthermore, to overcome multidrug resistance, nanoparticle-based systems have been developed due to their good therapeutic effect, low side effects, and high tumor metastasis inhibition. In view of this, we systematically discuss the molecular mechanisms and therapeutic strategies of MDR from nanotherapeutics. Finally, we summarize intriguing ideas and future trends for further research in overcoming MDR.
Collapse
Affiliation(s)
- Ayitila Maimaitijiang
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Dongze He
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Dingyang Li
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Wenfang Li
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Zhengding Su
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Zhongxiong Fan
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Jinyao Li
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| |
Collapse
|
6
|
Adu-Amankwaah F, Februarie C, Nyambo K, Maarman G, Tshililo N, Mabasa L, Mavumengwana V, Baatjies L. Cytotoxic properties, glycolytic effects and high-resolution respirometry mitochondrial activities of Eriocephalus racemosus against MDA-MB 231 triple-negative breast cancer. BMC Complement Med Ther 2024; 24:332. [PMID: 39256791 PMCID: PMC11389270 DOI: 10.1186/s12906-024-04615-x] [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: 05/17/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024] Open
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) represents a significant global health crisis due to its resistance to conventional therapies and lack of specific molecular targets. This study explored the potential of Eriocephalus racemosus (E. racemosus) as an alternative treatment for TNBC. The cytotoxic properties and high-resolution respirometry mitochondrial activities of E. racemosus against the MDA-MB 231 TNBC cell line were evaluated. METHODS Hexane solvent and bioactive fraction extractions of E. racemosus were performed, while mass spectrometry-based metabolite profiling was used to identify the phytochemical constituents of the extracts. The extracts were further tested against MDA-MB 231 TNBC cells to determine their cytotoxicity. The mode of cell death was determined using flow cytometry. The activities of caspases 3, 8, and 9 were assessed using a multiplex activity assay kit. Glycolytic activity and High-resolution respirometry measurements of mitochondrial function in the MDA-MB 231 cell line were conducted using the Seahorse XFp and Oroboros O2K. RESULTS Metabolite profiling of E. racemosus plant crude extracts identified the presence of coumarins, flavonoids, sesquiterpenoids, triterpenoids, and unknown compounds. The extracts demonstrated promising cytotoxic activities, with a half maximal inhibitory concentration (IC50) of 12.84 µg/mL for the crude hexane extract and 15.49 µg/mL for the bioactive fraction. Further, the crude hexane and bioactive fraction extracts induced apoptosis in the MDA-MB-231 TNBC cells, like the reference drug cisplatin (17.44%, 17.26% and 20.25%, respectively) compared to untreated cells. Caspase 3 activities confirmed the induction of apoptosis in both cisplatin and the plant crude extracts, while caspase 8 and 9 activities confirmed the activation of both the intrinsic and extrinsic apoptosis pathways. Increased levels of glycolytic activity were observed in the hexane crude extract. High-resolution respiratory measurements showed elevated mitochondrial activities in all mitochondrial states except for complex-IV activity. CONCLUSION These findings support further exploration of E. racemosus as a potential therapeutic agent for TNBC, offering a promising avenue for the development of targeted treatments with minimal adverse effects.
Collapse
Affiliation(s)
- Francis Adu-Amankwaah
- South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Candice Februarie
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health Science, CARMA: Centre for Cardio-Metabolic Research in Africa, Stellenbosch University, Cape Town, 8000, South Africa
| | - Kudakwashe Nyambo
- South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Gerald Maarman
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health Science, CARMA: Centre for Cardio-Metabolic Research in Africa, Stellenbosch University, Cape Town, 8000, South Africa
| | - Ndivhuwo Tshililo
- South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Lawrence Mabasa
- Biomedical Research and Innovation Platform (BRIP), Medical Research Council, Tygerberg, Cape Town, South Africa
| | - Vuyo Mavumengwana
- South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Biomedical Research and Innovation Platform (BRIP), Medical Research Council, Tygerberg, Cape Town, South Africa
| | - Lucinda Baatjies
- South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
- Biomedical Research and Innovation Platform (BRIP), Medical Research Council, Tygerberg, Cape Town, South Africa.
| |
Collapse
|
7
|
Yang F, Li S, Ji Q, Zhang H, Zhou M, Wang Y, Zhang S, Sun J, He Z, Luo C. Modular Prodrug-Engineered Oxygen Nano-Tank With Outstanding Nanoassembly Performance, High Oxygen Loading, and Closed-Loop Tumor Hypoxia Relief. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405583. [PMID: 38984484 PMCID: PMC11425644 DOI: 10.1002/advs.202405583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/30/2024] [Indexed: 07/11/2024]
Abstract
The clinical translation of tumor hypoxia intervention modalities still falls short of expectation, restricted by poor biocompatibility of oxygen-carrying materials, unsatisfactory oxygen loading performance, and abnormally high cellular oxygen consumption-caused insufficient hypoxia relief. Herein, a carrier-free oxygen nano-tank based on modular fluorination prodrug design and co-assembly nanotechnology is elaborately exploited, which is facilely fabricated through the molecular nanoassembly of a fluorinated prodrug (FSSP) of pyropheophorbide a (PPa) and an oxygen consumption inhibitor (atovaquone, ATO). The nano-tank adeptly achieves sufficient oxygen enrichment while simultaneously suppressing oxygen consumption within tumors for complete tumor hypoxia alleviation. Significant, the fluorination module in FSSP not only confers favorable co-assemblage of FSSP and ATO, but also empowers the nanoassembly to readily carry oxygen. As expected, it displays excellent oxygen carrying capacity, favorable pharmacokinetics, on-demand laser-triggerable ATO release, closed-loop tumor hypoxia relief, and significant enhancement to PPa-mediated PDT in vitro and in vivo. This study provides a novel nanotherapeutic paradigm for tumor hypoxia intervention-enhanced cancer therapy.
Collapse
Affiliation(s)
- Fujun Yang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Shumeng Li
- Department of Pharmaceutical Analysis, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Qingyu Ji
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Hongyuan Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Mingyang Zhou
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, 19104-6323, USA
| | - Yuequan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Shenwu Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| |
Collapse
|
8
|
Yu X, Xu C, Sun J, Xu H, Huang H, Gan Z, George A, Ouyang S, Liu F. Recent developments in two-dimensional molybdenum disulfide-based multimodal cancer theranostics. J Nanobiotechnology 2024; 22:515. [PMID: 39198894 PMCID: PMC11351052 DOI: 10.1186/s12951-024-02785-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/18/2024] [Indexed: 09/01/2024] Open
Abstract
Recent advancements in cancer research have led to the generation of innovative nanomaterials for improved diagnostic and therapeutic strategies. Despite the proven potential of two-dimensional (2D) molybdenum disulfide (MoS2) as a versatile platform in biomedical applications, few review articles have focused on MoS2-based platforms for cancer theranostics. This review aims to fill this gap by providing a comprehensive overview of the latest developments in 2D MoS2 cancer theranostics and emerging strategies in this field. This review highlights the potential applications of 2D MoS2 in single-model imaging and therapy, including fluorescence imaging, photoacoustic imaging, photothermal therapy, and catalytic therapy. This review further classifies the potential of 2D MoS2 in multimodal imaging for diagnostic and synergistic theranostic platforms. In particular, this review underscores the progress of 2D MoS2 as an integrated drug delivery system, covering a broad spectrum of therapeutic strategies from chemotherapy and gene therapy to immunotherapy and photodynamic therapy. Finally, this review discusses the current challenges and future perspectives in meeting the diverse demands of advanced cancer diagnostic and theranostic applications.
Collapse
Affiliation(s)
- Xinbo Yu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Shenyang, 110001, China
- Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Chen Xu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Shenyang, 110001, China
- Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Jingxu Sun
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Shenyang, 110001, China
| | - Hainan Xu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Hanwei Huang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Shenyang, 110001, China
- Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Ziyang Gan
- Institute of Physical Chemistry, Abbe Center of Photonics, Friedrich Schiller University Jena, Jena, Germany
| | - Antony George
- Institute of Physical Chemistry, Abbe Center of Photonics, Friedrich Schiller University Jena, Jena, Germany
| | - Sihui Ouyang
- College of Materials Science and Engineering, Chongqing University, National Engineering Research Center for Magnesium Alloys, Chongqing University, Chongqing, 400044, China.
| | - Funan Liu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Shenyang, 110001, China.
- Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
9
|
Chen W, Wang Z, Hong G, Du J, Song F, Peng X. Self-assembly-integrated tumor targeting and electron transfer programming towards boosting tumor type I photodynamic therapy. Chem Sci 2024; 15:10945-10953. [PMID: 39027272 PMCID: PMC11253188 DOI: 10.1039/d4sc03008g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/05/2024] [Indexed: 07/20/2024] Open
Abstract
Type I photodynamic therapy (PDT) is attracting increasing interest as an effective solution to the poor prognosis of patients with hypoxic tumors. The development of functional type I photosensitizers is limited by a lack of feasible strategies to systematically modulate electron transfer (ET) in photosensitization. Herein, we present an easily accessible approach for the preparation of nanophotosensitizers with self-assembly-integrated tumor-targeting and ET programming towards boosting tumor type I PDT. Specifically, a dual functional amphiphile PS-02 was designed with a ligand (6-NS) that had the ability to not only target tumor cell marker carbonic anhydrase IX (CAIX) but also regulate the ET process for type I PDT. The amphiphile PS-02 tended to self-assemble into PS-02 nanoparticles (NPs), which exhibited a local "ET-cage effect" due to the electron-deficient nature of 6-NS. It is noteworthy that when PS-02 NPs selectively targeted the tumor cells, the CAIX binding enabled the uncaging of the inhibited ET process owing to the electron-rich characteristic of CAIX. Therefore, PS-02 NPs integrated tumor targeting and CAIX activation towards boosting type I PDT. As a proof of concept, the improved PDT performance of PS-02 NPs was demonstrated with tumor cells under hypoxic conditions and solid tumor tissue in mouse in vivo experiments. This work provides a practical paradigm to develop versatile type I PDT nano-photosensitizers by simply manipulating ET and easy self-assembling.
Collapse
Affiliation(s)
- Wenlong Chen
- Shenzhen Research Institute of Shandong University, A301 Virtual University Park in South District of Shenzhen 518057 P. R. China
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology Dalian 116024 P. R. China
| | - Zehui Wang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology Dalian 116024 P. R. China
| | - Gaobo Hong
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology Dalian 116024 P. R. China
| | - Jianjun Du
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology Dalian 116024 P. R. China
| | - Fengling Song
- Shenzhen Research Institute of Shandong University, A301 Virtual University Park in South District of Shenzhen 518057 P. R. China
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology Dalian 116024 P. R. China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology Dalian 116024 P. R. China
| |
Collapse
|
10
|
Şahin Z, Önal E, Ali LMA, Durand D, Emami A, Touré M, İşci U, Gary-Bobo M, Cammas-Marion S, Dumoulin F. Nanoencapsulation of a Far-Red Absorbing Phthalocyanine into Poly(benzylmalate) Biopolymers and Modulation of Their Photodynamic Efficiency. Biomacromolecules 2024; 25:3261-3270. [PMID: 38752976 PMCID: PMC11170942 DOI: 10.1021/acs.biomac.3c01382] [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: 12/13/2023] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 06/11/2024]
Abstract
Two different poly(benzylmalate) biopolymers, a hydrophobic non-PEGylated (PMLABe73) and an amphiphilic PEGylated derivative (PEG42-b-PMLABe73), have been used to encapsulate a phthalocyanine chosen for its substitution pattern that is highly suitable for photodynamic therapy. Different phthalocyanine/(co)polymers ratios have been used for the nanoprecipitation. A set of six nanoparticles has been obtained. If the amphiphilic PEGylated copolymer proved to be slightly more efficient for the encapsulation and to lower the aggregation of the phthalocyanine inside the nanoparticles, it is, however, the hydrophobic PMLABe73-based nanoparticles that exhibited the best photodynamic efficiency.
Collapse
Affiliation(s)
- Zeynel Şahin
- Faculty
of Technology, Department of Metallurgical & Materials Engineering, Marmara University, 34722 Istanbul, Türkiye
| | - Emel Önal
- Faculty
of Engineering, Doğuş University, Ümraniye, 34775 Istanbul, Türkiye
| | - Lamiaa M. A. Ali
- IBMM,
Univ Montpellier, CNRS, ENSCM, 34093 Montpellier, France
- Department
of Biochemistry Medical Research Institute, University of Alexandria, 21561 Alexandria, Egypt
| | - Denis Durand
- IBMM,
Univ Montpellier, CNRS, ENSCM, 34093 Montpellier, France
| | - Atefeh Emami
- Faculty
of Engineering and Natural Sciences, Biomedical Engineering Department, Acıbadem Mehmet Ali Aydınlar University, Ataşehir, 34752 Istanbul, Türkiye
| | - Marie Touré
- IBMM,
Univ Montpellier, CNRS, ENSCM, 34093 Montpellier, France
| | - Umit İşci
- Faculty
of Technology, Department of Metallurgical & Materials Engineering, Marmara University, 34722 Istanbul, Türkiye
| | | | - Sandrine Cammas-Marion
- Univ
Rennes,
ENSCR, INSA Rennes, CNRS, ISCR (Institut des Sciences Chimiques de
Rennes)—UMR 6226, F-35000 Rennes, France
- INSERM,
INRAE, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer),
U1317, F-35000 Rennes, France
| | - Fabienne Dumoulin
- Faculty
of Engineering and Natural Sciences, Biomedical Engineering Department, Acıbadem Mehmet Ali Aydınlar University, Ataşehir, 34752 Istanbul, Türkiye
| |
Collapse
|
11
|
Zhi S, Huang M, Cheng K. Enzyme-responsive design combined with photodynamic therapy for cancer treatment. Drug Discov Today 2024; 29:103965. [PMID: 38552778 DOI: 10.1016/j.drudis.2024.103965] [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: 01/05/2024] [Revised: 03/09/2024] [Accepted: 03/22/2024] [Indexed: 04/07/2024]
Abstract
Photodynamic therapy (PDT) is a noninvasive cancer treatment that has garnered significant attention in recent years. However, its application is still hampered by certain limitations, such as the hydrophobicity and low targeting of photosensitizers (PSs) and the hypoxia of the tumor microenvironment. Nevertheless, the fusion of enzyme-responsive drugs with PDT offers novel solutions to overcome these challenges. Utilizing the attributes of enzyme-responsive drugs, PDT can deliver PSs to the target site and selectively release them, thereby enhancing therapeutic outcomes. In this review, we spotlight recent advances in enzyme-responsive materials for cancer treatment and primarily delineate their application in combination with PDT.
Collapse
Affiliation(s)
- Siying Zhi
- Guangdong Provincial Key Laboratory of New Drug Screening and NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Meixin Huang
- Guangdong Provincial Key Laboratory of New Drug Screening and NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Kui Cheng
- Guangdong Provincial Key Laboratory of New Drug Screening and NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
12
|
Ding Y, Yu W, Shen R, Zheng X, Zheng H, Yao Y, Zhang Y, Du C, Yi H. Hypoxia-Responsive Tetrameric Supramolecular Polypeptide Nanoprodrugs for Combination Therapy. Adv Healthc Mater 2024; 13:e2303308. [PMID: 37924332 DOI: 10.1002/adhm.202303308] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/30/2023] [Indexed: 11/06/2023]
Abstract
Despite the intense progress of photodynamic and chemotherapy, however, they cannot prevent solid tumor invasion, metastasis, and relapse, along with inferior efficacy and severe side effects. The hypoxia-responsive nanoprodrugs integrating photodynamic functions are highly sought to address the above-mentioned problems and overcome the tumor hypoxia-reduced efficacy. Herein, a hypoxia-responsive tetrameric supramolecular polypeptide nanoprodrug (SPN-TAPP-PCB4) is constructed from the self-assembly of tetrameric porphyrin-central poly(l-lysine-azobenzene-chlorambucil) (TAPP-(PLL-Azo-CB)4) and an anionic water-soluble [2]biphenyl-extended-pillar[6]arene (AWBpP6) via the synergy of hydrophobic, π-π stacking, and host-guest interactions. Upon laser irradiation, the central TAPP can convert oxygen to generate single oxygen (1 O2 ) to kill tumor cells. Furthermore, under the acidic and PDT-aggravated hypoxia tumor cell microenvironment, SPN-TAPP-PCB4 is rapidly disassembled, and then efficiently releases activated CB through the hypoxic-responsive cleavage of azobenzene linkages. Both in vitro and in vivo biological studies showcase synergistic cancer-killing actions between photodynamic therapy (PDT) and chemotherapy (CT) with negligible toxicity. Consequently, this supramolecular polypeptide nanoprodrug offers an effective strategy to design a hypoxia-responsive nanoprodrug for a potential combo PDT-CT transition.
Collapse
Affiliation(s)
- Yue Ding
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, 226019, P. R. China
| | - Wei Yu
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, 226019, P. R. China
| | - Rongkai Shen
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, 20, Chazhong Rd., Fuzhou, Fujian, 350005, China
| | - Xiangqin Zheng
- Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, National Key Clinical Specialty Construction Program of (Gynecology), Fujian Province Key Clinical Specialty for Gynecology, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, 350001, China
| | - Hui Zheng
- Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, National Key Clinical Specialty Construction Program of (Gynecology), Fujian Province Key Clinical Specialty for Gynecology, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, 350001, China
| | - Yong Yao
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, 226019, P. R. China
| | - Yuehua Zhang
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, 226019, P. R. China
| | - Chang Du
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Huan Yi
- Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, National Key Clinical Specialty Construction Program of (Gynecology), Fujian Province Key Clinical Specialty for Gynecology, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, 350001, China
| |
Collapse
|
13
|
Zhu X, Zheng W, Wang X, Li Z, Shen X, Chen Q, Lu Y, Chen K, Ai S, Zhu Y, Guan W, Yao S, Liu S. Enhanced Photodynamic Therapy Synergizing with Inhibition of Tumor Neutrophil Ferroptosis Boosts Anti-PD-1 Therapy of Gastric Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307870. [PMID: 38233204 DOI: 10.1002/advs.202307870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/19/2023] [Indexed: 01/19/2024]
Abstract
For tumor treatment, the ultimate goal in tumor therapy is to eliminate the primary tumor, manage potential metastases, and trigger an antitumor immune response, resulting in the complete clearance of all malignant cells. Tumor microenvironment (TME) refers to the local biological environment of solid tumors and has increasingly become an attractive target for cancer therapy. Neutrophils within TME of gastric cancer (GC) spontaneously undergo ferroptosis, and this process releases oxidized lipids that limit T cell activity. Enhanced photodynamic therapy (PDT) mediated by di-iodinated IR780 (Icy7) significantly increases the production of reactive oxygen species (ROS). Meanwhile, neutrophil ferroptosis can be triggered by increased ROS generation in the TME. In this study, a liposome encapsulating both ferroptosis inhibitor Liproxstatin-1 and modified photosensitizer Icy7, denoted LLI, significantly inhibits tumor growth of GC. LLI internalizes into MFC cells to generate ROS causing immunogenic cell death (ICD). Simultaneously, liposome-deliver Liproxstatin-1 effectively inhibits the ferroptosis of tumor neutrophils. LLI-based immunogenic PDT and neutrophil-targeting immunotherapy synergistically boost the anti-PD-1 treatment to elicit potent TME and systemic antitumor immune response with abscopal effects. In conclusion, LLI holds great potential for GC immunotherapy.
Collapse
Affiliation(s)
- Xudong Zhu
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Wenxuan Zheng
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Xingzhou Wang
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Zhiyan Li
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Xiaofei Shen
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Qi Chen
- China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, 210008, China
| | - Yanjun Lu
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Kai Chen
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Shichao Ai
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Yun Zhu
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Wenxian Guan
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Shankun Yao
- State Key Laboratory of Coordination Chemistry, Coordination Chemistry Institute, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Song Liu
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| |
Collapse
|
14
|
Mahapatra S, Ma S, Dong B, Zhang C. Quantification of cellular phototoxicity of organelle stains by the dynamics of microtubule polymerization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.17.576021. [PMID: 38293099 PMCID: PMC10827188 DOI: 10.1101/2024.01.17.576021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Being able to quantify the phototoxicity of dyes and drugs in live cells allows biologists to better understand cell responses to exogenous stimuli during imaging. This capability further helps to design fluorescent labels with lower phototoxicity and drugs with better efficacy. Conventional ways to evaluate cellular phototoxicity rely on late-stage measurements of individual or different populations of cells. Here, we developed a quantitative method using intracellular microtubule polymerization as a rapid and sensitive marker to quantify early-stage phototoxicity. Implementing this method, we assessed the photosensitization induced by organelle dyes illuminated with different excitation wavelengths. Notably, fluorescent markers targeting mitochondria, nuclei, and endoplasmic reticulum exhibited diverse levels of phototoxicity. Furthermore, leveraging a real-time precision opto-control technology allowed us to evaluate the synergistic effect of light and dyes on specific organelles. Studies in hypoxia revealed enhanced phototoxicity of Mito-Tracker Red CMXRos that is not correlated with the generation of reactive oxygen species but a different deleterious pathway in low oxygen conditions. Teaser Microtubule dynamics in live cells allow quantification of cellular phototoxicity of fluorescent dyes in various conditions.
Collapse
|
15
|
Xue Q, Zeng S, Ren Y, Pan Y, Chen J, Chen N, Wong KKY, Song L, Fang C, Guo J, Xu J, Liu C, Zeng J, Sun L, Zhang H, Chen J. Relief of tumor hypoxia using a nanoenzyme amplifies NIR-II photoacoustic-guided photothermal therapy. BIOMEDICAL OPTICS EXPRESS 2024; 15:59-76. [PMID: 38223179 PMCID: PMC10783917 DOI: 10.1364/boe.499286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 11/28/2023] [Accepted: 11/28/2023] [Indexed: 01/16/2024]
Abstract
Hypoxia is a critical tumor microenvironment (TME) component. It significantly impacts tumor growth and metastasis and is known to be a major obstacle for cancer therapy. Integrating hypoxia modulation with imaging-based monitoring represents a promising strategy that holds the potential for enhancing tumor theranostics. Herein, a kind of nanoenzyme Prussian blue (PB) is synthesized as a metal-organic framework (MOF) to load the second near-infrared (NIR-II) small molecule dye IR1061, which could catalyze hydrogen peroxide to produce oxygen and provide a photothermal conversion element for photoacoustic imaging (PAI) and photothermal therapy (PTT). To enhance stability and biocompatibility, silica was used as a coating for an integrated nanoplatform (SPI). SPI was found to relieve the hypoxic nature of the TME effectively, thus suppressing tumor cell migration and downregulating the expression of heat shock protein 70 (HSP70), both of which led to an amplified NIR-II PTT effect in vitro and in vivo, guided by the NIR-II PAI. Furthermore, label-free multi-spectral PAI permitted the real-time evaluation of SPI as a putative tumor treatment. A clinical histological analysis confirmed the amplified treatment effect. Hence, SPI combined with PAI could offer a new approach for tumor diagnosing, treating, and monitoring.
Collapse
Affiliation(s)
- Qiang Xue
- Department of Ultrasound, Shenzhen People's Hospital, The Second Clinical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518020, China
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Silue Zeng
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yaguang Ren
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yingying Pan
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Department of Medical Ultrasonics, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jianhai Chen
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ningbo Chen
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- The University of Hong Kong, Department of Electrical and Electronic Engineering, Hong Kong, China
| | - Kenneth K Y Wong
- The University of Hong Kong, Department of Electrical and Electronic Engineering, Hong Kong, China
| | - Liang Song
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Chihua Fang
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jinhan Guo
- Department of Ultrasound, Shenzhen People's Hospital, The Second Clinical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518020, China
| | - Jinfeng Xu
- Department of Ultrasound, Shenzhen People's Hospital, The Second Clinical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518020, China
| | - Chengbo Liu
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jie Zeng
- Department of Medical Ultrasonics, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Litao Sun
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Hai Zhang
- Department of Ultrasound, Shenzhen People's Hospital, The Second Clinical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518020, China
| | - Jingqin Chen
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
16
|
Aebisher D, Woźnicki P, Dynarowicz K, Kawczyk-Krupka A, Cieślar G, Bartusik-Aebisher D. Photodynamic Therapy and Immunological View in Gastrointestinal Tumors. Cancers (Basel) 2023; 16:66. [PMID: 38201494 PMCID: PMC10777986 DOI: 10.3390/cancers16010066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Gastrointestinal cancers are a specific group of oncological diseases in which the location and nature of growth are of key importance for clinical symptoms and prognosis. At the same time, as research shows, they pose a serious threat to a patient's life, especially at an advanced stage of development. The type of therapy used depends on the anatomical location of the cancer, its type, and the degree of progression. One of the modern forms of therapy used to treat gastrointestinal cancers is PDT, which has been approved for the treatment of esophageal cancer in the United States. Despite the increasingly rapid clinical use of this treatment method, the exact immunological mechanisms it induces in cancer cells has not yet been fully elucidated. This article presents a review of the current understanding of the mode of action of photodynamic therapy on cells of various gastrointestinal cancers with an emphasis on colorectal cancer. The types of cell death induced by PDT include apoptosis, necrosis, and pyroptosis. Anticancer effects are also a result of the destruction of tumor vasculature and activation of the immune system. Many reports exist that concern the mechanism of apoptosis induction, of which the mitochondrial pathway is most often emphasized. Photodynamic therapy may also have a beneficial effect on such aspects of cancer as the ability to develop metastases or contribute to reducing resistance to known pharmacological agents.
Collapse
Affiliation(s)
- David Aebisher
- Department of Photomedicine and Physical Chemistry, Medical College of the University of Rzeszów, 35-959 Rzeszów, Poland
| | - Paweł Woźnicki
- Students English Division Science Club, Medical College of the University of Rzeszów, 35-959 Rzeszów, Poland
| | - Klaudia Dynarowicz
- Center for Innovative Research in Medical and Natural Sciences, Medical College of the University of Rzeszów, 35-310 Rzeszów, Poland;
| | - Aleksandra Kawczyk-Krupka
- Department of Internal Medicine, Angiology and Physical Medicine, Center for Laser Diagnostics and Therapy, Medical University of Silesia, Batorego 15 Street, 41-902 Bytom, Poland; (A.K.-K.); (G.C.)
| | - Grzegorz Cieślar
- Department of Internal Medicine, Angiology and Physical Medicine, Center for Laser Diagnostics and Therapy, Medical University of Silesia, Batorego 15 Street, 41-902 Bytom, Poland; (A.K.-K.); (G.C.)
| | - Dorota Bartusik-Aebisher
- Department of Biochemistry and General Chemistry, Medical College of the University of Rzeszów, 35-959 Rzeszów, Poland;
| |
Collapse
|
17
|
Sharif S, Jung D, Cao HX, Park JO, Kang B, Choi E. Ultrasonic Manipulation of Hydrodynamically Driven Microparticles in Vessel Bifurcation: Simulation, Optimization, Experimental Validation, and Potential for Targeted Drug Delivery. MICROMACHINES 2023; 15:13. [PMID: 38276841 PMCID: PMC10819303 DOI: 10.3390/mi15010013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024]
Abstract
Microrobots driven by multiple external power sources have emerged as promising tools for targeted drug and stem cell delivery in tissue regeneration. However, navigating and imaging these devices within a complex colloidal vascular system at a clinical scale is challenging. Ultrasonic actuators have gained interest in the field of non-contact manipulation of micromachines due to their label-free biocompatible nature and safe operation history. This research presents experimentally validated simulation results of ultrasonic actuation using a novel ultrasonic transducer array with a hemispherical arrangement that generates active traveling waves with phase modulation. Blood flow is used as a carrier force while the direction and path are controlled by blocking undesirable paths using a highly focused acoustic field. In the experiments, the microrobot cluster was able to follow a predefined trajectory and reach the target. The microrobot size, maximum radiation pressure, and focus position were optimized for certain blood flow conditions. The outcomes suggest that this acoustic manipulation module has potential applications in targeted tumor therapy.
Collapse
Affiliation(s)
- Saqib Sharif
- School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea;
- Korea Institute of Medical Microrobotics, Gwangju 61011, Republic of Korea; (D.J.); (H.X.C.); (J.-O.P.)
| | - Daewon Jung
- Korea Institute of Medical Microrobotics, Gwangju 61011, Republic of Korea; (D.J.); (H.X.C.); (J.-O.P.)
| | - Hiep Xuan Cao
- Korea Institute of Medical Microrobotics, Gwangju 61011, Republic of Korea; (D.J.); (H.X.C.); (J.-O.P.)
- College of AI Convergence, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jong-Oh Park
- Korea Institute of Medical Microrobotics, Gwangju 61011, Republic of Korea; (D.J.); (H.X.C.); (J.-O.P.)
| | - Byungjeon Kang
- Korea Institute of Medical Microrobotics, Gwangju 61011, Republic of Korea; (D.J.); (H.X.C.); (J.-O.P.)
- College of AI Convergence, Chonnam National University, Gwangju 61186, Republic of Korea
- Graduate School of Data Science, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Eunpyo Choi
- School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea;
- Korea Institute of Medical Microrobotics, Gwangju 61011, Republic of Korea; (D.J.); (H.X.C.); (J.-O.P.)
- College of AI Convergence, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
18
|
Lei L, Dai W, Man J, Hu H, Jin Q, Zhang B, Tang Z. Lonidamine liposomes to enhance photodynamic and photothermal therapy of hepatocellular carcinoma by inhibiting glycolysis. J Nanobiotechnology 2023; 21:482. [PMID: 38102658 PMCID: PMC10724989 DOI: 10.1186/s12951-023-02260-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/11/2023] [Indexed: 12/17/2023] Open
Abstract
Phototherapy, including photodynamic therapy (PDT) and photothermal therapy (PTT), has great promise in the treatment of cancer. However, there are many obstacles that can restrict the therapeutic efficacy of phototherapy. The hypoxic tumor microenvironment can restrict the production of reactive oxygen species (ROS) in PDT. As for PTT, the thermotolerance of cancer cells may lead to ineffective PTT. In this study, IR780 and glycolysis inhibitor lonidamine (LND)-encapsulated liposomes are prepared for photodynamic and photothermal therapy of hepatocellular carcinoma. IR780 can be used as a photosensitizer and photothermal agent for simultaneous PDT and PTT after being irradiated with 808 nm laser. LND can reduce the oxygen consumption of cancer cells by inhibiting glycolysis, which will relieve tumor hypoxia and produce more ROS for PDT. On the other hand, energy supply can be blocked by LND-induced glycolysis inhibition, which will inhibit the production of heat shock proteins (HSPs), reduce the thermotolerance of tumor cells, and finally enhance the therapeutic efficacy of PTT. The enhanced PTT is studied by measuring intracellular HSPs, ATP level, and mitochondrial membrane potential. The antitumor effect of IR780 and LND co-loaded liposomes is extensively investigated by in vitro and in vivo experiments. This research provides an innovative strategy to simultaneously enhance the therapeutic efficacy of PDT and PTT by inhibiting glycolysis, which is promising for future creative approaches to cancer phototherapy.
Collapse
Affiliation(s)
- Lei Lei
- Department of Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Wenbin Dai
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jiaping Man
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Haitao Hu
- Department of Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Qiao Jin
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Bo Zhang
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Zhe Tang
- Department of Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, 322000, China.
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
19
|
Yang C, Tian S, Qiu W, Mo L, Lin W. Hierarchical MOF@AuNP/Hairpin Nanotheranostic for Enhanced Photodynamic Therapy via O 2 Self-Supply and Cancer-Related MicroRNA Imaging In Vivo. Anal Chem 2023; 95:16279-16288. [PMID: 37870556 DOI: 10.1021/acs.analchem.3c03216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Developing a nanotheranostic with a high sensing performance and efficient therapy was significant in cancer diagnosis and treatment. Herein, a Au nanoparticle and hairpin-loaded photosensitive metal-organic framework (PMOF@AuNP/hairpin) nanotheranostic was constructed by growing AuNPs on PMOF in situ and then attaching hairpins. On the one hand, the PMOF@AuNP/hairpin nanotheranostic could effectively transfer O2 into ROS, facilitating efficient PDT. Additionally, the nanotheranostic possessed catalase-like activity, which could effectively catalyze H2O2 to generate O2, thus achieving O2-evolving PDT and significantly enhancing the antitumor effect of PDT in vivo. On the other hand, the nanotheranostic showed a high loading efficiency of hairpins and achieved the sensitive and selective detection of miR-21 both in living cells and in vivo. Moreover, the nanotheranostic could dynamically monitor the miR-21 level. Due to the excellent imaging performance, the nanotheranostic could recognize cancer cells and might provide important information on cancer progression for PDT. The developed PMOF@AuNP/hairpin nanotheranostic provided a useful tool for tumor diagnosis and antitumor therapy.
Collapse
Affiliation(s)
- Chan Yang
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi 530004, P. R. China
| | - Shuo Tian
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi 530004, P. R. China
| | - Weiyu Qiu
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi 530004, P. R. China
| | - Liuting Mo
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi 530004, P. R. China
| | - Weiying Lin
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi 530004, P. R. China
| |
Collapse
|
20
|
Ma S, Xu W, Fei Y, Li D, Jia X, Wang J, Wang E. Mn 2+ /Ir 3+ -Doped and CaCO 3 -Covered Prussian Blue Nanoparticles with Indocyanine Green Encapsulation for Tumor Microenvironment Modulation and Image-Guided Synergistic Cancer Therapy. Adv Healthc Mater 2023; 12:e2301413. [PMID: 37657182 DOI: 10.1002/adhm.202301413] [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: 05/04/2023] [Revised: 08/20/2023] [Indexed: 09/03/2023]
Abstract
The development of smart theranostic nanoplatforms has gained great interest in effective cancer treatment against the complex tumor microenvironment (TME), including weak acidity, hypoxia, and glutathione (GSH) overexpression. Herein, a TME-responsive nanoplatform named PMICApt /ICG, based on PB:Mn&Ir@CaCO3 Aptamer /ICG, is designed for the competent synergistic photothermal therapy and photodynamic therapy (PDT) under the guidance of photothermal and magnetic resonance imaging. The nanoplatform's aptamer modification targeting the transferrin receptor and the epithelial cell adhesion molecule on breast cancer cells, and the acid degradable CaCO3 shell allow for effective tumor accumulation and TME-responsive payload release in situ. The nanoplatform also exhibits excellent PDT properties due to its ability to generate O2 and consume antioxidant GSH in tumors. Additionally, the synergistic therapy is achieved by a single wavelength of near-infrared laser. RNA sequencing is performed to identify differentially expressed genes, which show that the expressions of proliferation and migration-associated genes are inhibited, while the apoptosis and immune response gene expressions are upregulated after the synergistic treatments. This multifunctional nanoplatform that responds to the TME to realize the on-demand payload release and enhance PDT induced by TME modulation holds great promise for clinical applications in tumor therapy.
Collapse
Affiliation(s)
- Shuaining Ma
- College of Physics, Jilin University, Changchun, Jilin, 130012, P. R. China
- State Key Laboratory of Electroanalytical Chemistry, Key Laboratory of Polymer Ecomaterials (W. Xu), Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
| | - Weiguo Xu
- State Key Laboratory of Electroanalytical Chemistry, Key Laboratory of Polymer Ecomaterials (W. Xu), Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
| | - Yunwei Fei
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, 130012, P. R. China
| | - Dan Li
- State Key Laboratory of Electroanalytical Chemistry, Key Laboratory of Polymer Ecomaterials (W. Xu), Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
| | - Xiuna Jia
- State Key Laboratory of Electroanalytical Chemistry, Key Laboratory of Polymer Ecomaterials (W. Xu), Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
| | - Jin Wang
- Department of Chemistry and Physics, State University of New York at Stony Brook, Stony Brook, NY, 11794-3400, USA
| | - Erkang Wang
- State Key Laboratory of Electroanalytical Chemistry, Key Laboratory of Polymer Ecomaterials (W. Xu), Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
| |
Collapse
|
21
|
Liu K, Yao Y, Xue S, Zhang M, Li D, Xu T, Zhi F, Liu Y, Ding D. Recent Advances of Tumor Microenvironment-Responsive Nanomedicines-Energized Combined Phototherapy of Cancers. Pharmaceutics 2023; 15:2480. [PMID: 37896240 PMCID: PMC10610502 DOI: 10.3390/pharmaceutics15102480] [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: 08/28/2023] [Revised: 10/02/2023] [Accepted: 10/08/2023] [Indexed: 10/29/2023] Open
Abstract
Photodynamic therapy (PDT) has emerged as a powerful tumor treatment tool due to its advantages including minimal invasiveness, high selectivity and thus dampened side effects. On the other side, the efficacy of PDT is severely frustrated by the limited oxygen level in tumors, thus promoting its combination with other therapies, particularly photothermal therapy (PTT) for bolstered tumor treatment outcomes. Meanwhile, nanomedicines that could respond to various stimuli in the tumor microenvironment (TME) provide tremendous benefits for combined phototherapy with efficient hypoxia relief, tailorable drug release and activation, improved cellular uptake and intratumoral penetration of nanocarriers, etc. In this review, we will introduce the merits of combining PTT with PDT, summarize the recent important progress of combined phototherapies and their combinations with the dominant tumor treatment regimen, chemotherapy based on smart nanomedicines sensitive to various TME stimuli with a focus on their sophisticated designs, and discuss the challenges and future developments of nanomedicine-mediated combined phototherapies.
Collapse
Affiliation(s)
- Kehan Liu
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; (K.L.); (S.X.); (M.Z.); (T.X.)
| | - Yao Yao
- Department of Gerontology, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian 223800, China;
| | - Shujuan Xue
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; (K.L.); (S.X.); (M.Z.); (T.X.)
| | - Mengyao Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; (K.L.); (S.X.); (M.Z.); (T.X.)
| | - Dazhao Li
- Department of Neurosurgery, The First People’s Hospital of Changzhou, Changzhou 213003, China; (D.L.); (F.Z.)
- Clinical Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Tao Xu
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; (K.L.); (S.X.); (M.Z.); (T.X.)
- School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), D02 NY74 Dublin, Ireland
| | - Feng Zhi
- Department of Neurosurgery, The First People’s Hospital of Changzhou, Changzhou 213003, China; (D.L.); (F.Z.)
- Clinical Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Yang Liu
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; (K.L.); (S.X.); (M.Z.); (T.X.)
| | - Dawei Ding
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; (K.L.); (S.X.); (M.Z.); (T.X.)
| |
Collapse
|
22
|
Jo S, Lee H, Park JH, Yang JK, Lee WJ, Lim J, Kim S, Lee S, Lee TS. Silica-Based Platform Decorated with Conjugated Polymer Dots and Prussian Blue for Improved Photodynamic Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:43455-43467. [PMID: 37682242 DOI: 10.1021/acsami.3c08404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
To advance cancer treatment, we have developed a novel composite material consisting of conjugated polymer dots (CPDs) and Prussian blue (PB) particles, which were immobilized on, and encapsulated within, silica particles, respectively. The CPDs functioned as both a photosensitizer and a photodynamic agent, and the PB acted as a photothermal agent. The silica platform provided a biocompatible matrix that brought the two components into close proximity. Under laser irradiation, the fluorescence from the CPDs in the composite material enabled cell imaging and was subsequently converted to thermal energy by PB. This efficient energy transfer was accomplished because of the spectral overlap between the emission of donor CPDs and the absorbance of acceptor PB. The increase in local temperature in the cells resulted in a significant increase in the amount of reactive oxygen species (ROS) generated by CPDs, in which their independent use did not produce sufficient ROS for cancer cell treatment. To assess the impact of the enhanced ROS generation by the composite material, we conducted experiments using cancer cells under 532 nm laser irradiation. The results showed that with the increase in local temperature, the generated ROS increased by 30% compared with the control, which did not contain PB. When the silica-based composite material was positioned at the periphery of the tumor for 120 h, it led to a much slower tumor growth than other materials tested. By using a CPD-based photodynamic therapy platform, a new simplified approach to designing and preparing cancer treatments could be achieved, which included photothermal PB-assisted enhanced ROS generation using a single laser. This advancement opens up an exciting new opportunity for effective cancer treatment.
Collapse
Affiliation(s)
- Seonyoung Jo
- Department of Organic Materials Engineering, Chungnam National University, Daejeon 34134, Korea
| | - Hyeonhee Lee
- Department of Microbiology & Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Ji Hwan Park
- Center for Theragnosis, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Jin-Kyoung Yang
- Center for Theragnosis, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Won-Jong Lee
- Graduate School of Energy Science and Technology, Chungnam National University, Daejeon 34134, Korea
| | - Jongchul Lim
- Graduate School of Energy Science and Technology, Chungnam National University, Daejeon 34134, Korea
| | - Sehoon Kim
- Center for Theragnosis, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Soojin Lee
- Department of Microbiology & Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Taek Seung Lee
- Department of Organic Materials Engineering, Chungnam National University, Daejeon 34134, Korea
| |
Collapse
|
23
|
Wu Q, Ma Q, Ma J, Chen J, Zhuang B, Yang S, Liu J, Wen S. Cascade Amplification of Pyroptosis and Apoptosis for Cancer Therapy through a Black Phosphorous-Doped Thermosensitive Hydrogel. Pharmaceutics 2023; 15:1830. [PMID: 37514017 PMCID: PMC10383820 DOI: 10.3390/pharmaceutics15071830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/22/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
Cell pyroptosis has a reciprocal relationship with various cancer treatment modalities such as chemotherapy. However, the tumor microenvironment, characterized by hypoxia, substantially restricts the development and application of tumor therapies that integrate cell pyroptosis. Therefore, the cascade amplification of oxidative stress by interfering with redox homeostasis in tumors may be a promising approach. In this study, black phosphorus (BP) nanosheets and a glutathione peroxidase 4 inhibitor (RSL3) were coloaded into a thermosensitive PDLLA-PEG-PDLLA (PLEL) hydrogel (RSL3/BP@PLEL). Owing to the photothermal property of BP nanosheets, the RSL3/BP@PLEL hydrogel may trigger the release of loaded drugs in a more controllable and on-demand manner. Investigation of the antitumor effect in a mouse liver tumor model demonstrated that local injection of the hydrogel formulation in combination with near infrared laser irradiation could efficiently suppress tumor growth by interfering with the redox balance in tumors. Mechanistic study indicated that the combined treatment of photothermal therapy and glutathione depletion based on this hydrogel efficiently induced cell pyroptosis through both caspase-1/GSDMD and caspase-3/GSDME pathways, thereby triggering the repolarization of tumor-associated macrophages from M2 to M1. Overall, we developed a biocompatible and biodegradable hydrogel formulation for application in combination cancer treatment, providing a new platform for enhancing the efficacy of cancer therapy by amplifying cell pyroptosis and apoptosis.
Collapse
Affiliation(s)
- Qing Wu
- Department of Hepatic-Biliary-Pancreatic Surgery, Affiliated Foshan Hospital of Southern Medical University, Foshan 528000, China
| | - Qinghui Ma
- Department of Oncology, Affiliated Foshan Hospital of Southern Medical University, Foshan 528000, China
| | - Jun Ma
- Department of Gastroenterology, Affiliated Foshan Hospital of Southern Medical University, Foshan 528000, China
| | - Junpeng Chen
- Department of Hepatic-Biliary-Pancreatic Surgery, Affiliated Foshan Hospital of Southern Medical University, Foshan 528000, China
| | - Baoding Zhuang
- Department of Hepatic-Biliary-Pancreatic Surgery, Affiliated Foshan Hospital of Southern Medical University, Foshan 528000, China
| | - Shanglin Yang
- Department of Hepatic-Biliary-Pancreatic Surgery, Affiliated Foshan Hospital of Southern Medical University, Foshan 528000, China
| | - Jinji Liu
- Department of Oncology, Affiliated Foshan Hospital of Southern Medical University, Foshan 528000, China
| | - Shunqian Wen
- Department of Hepatic-Biliary-Pancreatic Surgery, Affiliated Foshan Hospital of Southern Medical University, Foshan 528000, China
| |
Collapse
|
24
|
Tran TH, Tran PTT, Truong DH. Lactoferrin and Nanotechnology: The Potential for Cancer Treatment. Pharmaceutics 2023; 15:pharmaceutics15051362. [PMID: 37242604 DOI: 10.3390/pharmaceutics15051362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/12/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Lactoferrin (Lf)-a glycoprotein of the transferrin family-has been investigated as a promising molecule with diverse applications, including infection inhibition, anti-inflammation, antioxidant properties and immune modulation. Along with that, Lf was found to inhibit the growth of cancerous tumors. Owing to unique properties such as iron-binding and positive charge, Lf could interrupt the cancer cell membrane or influence the apoptosis pathway. In addition, being a common mammalian excretion, Lf offers is promising in terms of targeting delivery or the diagnosis of cancer. Recently, nanotechnology significantly enhanced the therapeutic index of natural glycoproteins such as Lf. Therefore, in the context of this review, the understanding of Lf is summarized and followed by different strategies of nano-preparation, including inorganic nanoparticles, lipid-based nanoparticles and polymer-based nanoparticles in cancer management. At the end of the study, the potential future applications are discussed to pave the way for translating Lf into actual usage.
Collapse
Affiliation(s)
- Tuan Hiep Tran
- Faculty of Pharmacy, Phenikaa University, Yen Nghia, Ha Dong, Hanoi 12116, Vietnam
| | - Phuong Thi Thu Tran
- Department of Life Sciences, University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi 10000, Vietnam
| | | |
Collapse
|
25
|
Zhang Y, Wang R, Liu R, Xie S, Jiao F, Li Y, Xin J, Zhang H, Wang Z, Yan Y. Delivery of miR-3529-3p using MnO 2 -SiO 2 -APTES nanoparticles combined with phototherapy suppresses lung adenocarcinoma progression by targeting HIGD1A. Thorac Cancer 2023; 14:913-928. [PMID: 36808485 PMCID: PMC10067359 DOI: 10.1111/1759-7714.14823] [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: 12/25/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND The present study aimed to investigate the function of miR-3529-3p in lung adenocarcinoma and MnO2 -SiO2 -APTES (MSA) as a promising multifunctional delivery agent for lung adenocarcinoma therapy. METHODS Expression levels of miR-3529-3p were evaluated in lung carcinoma cells and tissues by qRT-PCR. The effects of miR-3529-3p on apoptosis, proliferation, metastasis and neovascularization were assessed by CCK-8, FACS, transwell and wound healing assays, tube formation and xenografts experiments. Luciferase reporter assays, western blot, qRT-PCR and mitochondrial complex assay were used to determine the targeting relationship between miR-3529-3p and hypoxia-inducible gene domain family member 1A (HIGD1A). MSA was fabricated using MnO2 nanoflowers, and its heating curves, temperature curves, IC50, and delivery efficiency were examined. The hypoxia and reactive oxygen species (ROS) production was investigated by nitro reductase probing, DCFH-DA staining and FACS. RESULTS MiR-3529-3p expression was reduced in lung carcinoma tissues and cells. Transfection of miR-3529-3p could promote apoptosis and suppress cell proliferation, migration and angiogenesis. As a target of miR-3529-3p, HIGD1A expression was downregulated, through which miR-3529-3p could disrupt the activities of complexes III and IV of the respiratory chain. The multifunctional nanoparticle MSA could not only efficiently deliver miR-3529-3p into cells, but also enhance the antitumor function of miR-3529-3p. The underlying mechanism may be that MSA alleviates hypoxia and has synergistic effects in cellular ROS promotion with miR-3529-3p. CONCLUSIONS Our results establish the antioncogenic role of miR-3529-3p, and demonstrate that miR-3529-3p delivered by MSA has enhanced tumor suppressive effects, probably through elevating ROS production and thermogenesis.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiP. R. China
- Oncology DepartmentBinzhou Medical University HospitalBinzhouP. R. China
| | - Ran‐Ran Wang
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiP. R. China
| | - Rui Liu
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiP. R. China
| | - Shu‐Yang Xie
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiP. R. China
| | - Fei Jiao
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiP. R. China
| | - You‐Jie Li
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiP. R. China
| | - Jiaxuan Xin
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiP. R. China
| | - Han Zhang
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiP. R. China
| | - Zhenbo Wang
- Oncology DepartmentBinzhou Medical University HospitalBinzhouP. R. China
| | - Yun‐Fei Yan
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiP. R. China
| |
Collapse
|
26
|
Zhang XH, Song YC, Qiu F, Wang ZC, Li N, Zhao FB. Hypoxic glioma cell-secreted exosomal circ101491 promotes the progression of glioma by regulating miR-125b-5p/EDN1. Brain Res Bull 2023; 195:55-65. [PMID: 36796652 DOI: 10.1016/j.brainresbull.2023.02.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/02/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023]
Abstract
Hypoxia and exosomes play important roles in the occurrence and development of glioma. While circRNAs are involved in biological processes of various tumors, the mechanism underlying exosome-dependent regulatory effects of circRNAs on the progression of glioma under hypoxia is unclear. Results suggested that circ101491 was overexpressed in tumor tissues and plasma exosomes of glioma patients, while the overexpression of circ101491 was closely related to the differentiation degree and TNM staging of the patients. Moreover, circ101491 overexpression promoted viability, invasion and migration of glioma cells both in vivo and in vitro; the above regulatory effects can be reversed by inhibition of circ101491 expression. Mechanistic studies revealed that circ101491 upregulated EDN1 expression through sponging miR-125b-5p, thus facilitating glioma progression. In summary, hypoxia could promote circ101491 overexpression in glioma cell-derived exosomes, and circ101491/miR-125b-5p/EDN1 regulatory axis might be implicated in the malignant progression of glioma.
Collapse
Affiliation(s)
- Xiao-Hui Zhang
- Department of Pathology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, PR China.
| | - Yi-Cun Song
- Department of Pathology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Feng Qiu
- Department of Pathology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Zheng-Cai Wang
- Department of Pathology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Nan Li
- Department of Pathology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, PR China
| | - Fang-Bo Zhao
- College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, PR China
| |
Collapse
|
27
|
Schneider L, Kalt M, Koch S, Sithamparanathan S, Villiger V, Mattiat J, Kradolfer F, Slyshkina E, Luber S, Bonmarin M, Maake C, Spingler B. BODIPY-Based Photothermal Agents with Excellent Phototoxic Indices for Cancer Treatment. J Am Chem Soc 2023; 145:4534-4544. [PMID: 36780327 DOI: 10.1021/jacs.2c11650] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Here, we report six novel, easily accessible BODIPY-based agents for cancer treatment. In contrast to established photodynamic therapy (PDT) agents, these BODIPY-based compounds show additional photothermal activity and their cytotoxicity is not dependent on the generation of reactive oxygen species (ROS). The agents show high photocytotoxicity upon irradiation with light and low dark toxicity in different cancer cell lines in 2D culture as well as in 3D multicellular tumor spheroids (MCTSs). The ratio of dark to light toxicity (phototoxic index, PI) of these agents reaches striking values exceeding 830,000 after irradiation with energetically low doses of light at 630 nm. The oxygen-dependent mechanism of action (MOA) of established photosensitizers (PSs) hampers effective clinical deployment of these agents. Under hypoxic conditions (0.2% O2), which are known to limit the efficiency of conventional PSs in solid tumors, photocytotoxicity was induced at the same concentration levels, indicating an oxygen-independent photothermal MOA. With a PI exceeding 360,000 under hypoxic conditions, both PI values are the highest reported to date. We anticipate that small molecule agents with a photothermal MOA, such as the BODIPY-based compounds reported in this work, may overcome this barrier and provide a new avenue to cancer therapy.
Collapse
Affiliation(s)
- Lukas Schneider
- Department of Chemistry, University of Zurich, CH-8057 Zurich, Switzerland
| | - Martina Kalt
- Department of Chemistry, University of Zurich, CH-8057 Zurich, Switzerland.,Institute of Anatomy, University of Zurich, CH-8057 Zurich, Switzerland
| | - Samuel Koch
- Department of Chemistry, University of Zurich, CH-8057 Zurich, Switzerland
| | | | - Veronika Villiger
- Department of Chemistry, University of Zurich, CH-8057 Zurich, Switzerland
| | - Johann Mattiat
- Department of Chemistry, University of Zurich, CH-8057 Zurich, Switzerland
| | - Flavia Kradolfer
- Department of Chemistry, University of Zurich, CH-8057 Zurich, Switzerland
| | | | - Sandra Luber
- Department of Chemistry, University of Zurich, CH-8057 Zurich, Switzerland
| | - Mathias Bonmarin
- School of Engineering, Zurich University of Applied Sciences, CH-8400 Winterthur, Switzerland
| | - Caroline Maake
- Institute of Anatomy, University of Zurich, CH-8057 Zurich, Switzerland
| | - Bernhard Spingler
- Department of Chemistry, University of Zurich, CH-8057 Zurich, Switzerland
| |
Collapse
|
28
|
Hu H, Xu D, Xu Q, Tang Y, Hong J, Hu Y, Wang J, Ni X. Reduction-responsive worm-like nanoparticles for synergistic cancer chemo-photodynamic therapy. Mater Today Bio 2023; 18:100542. [PMID: 36647538 PMCID: PMC9840183 DOI: 10.1016/j.mtbio.2023.100542] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/28/2022] [Accepted: 01/03/2023] [Indexed: 01/05/2023] Open
Abstract
Chemo-photodynamic therapy shows great potential for cancer treatment. However, the rational integration of chemotherapeutic agents and photosensitizers to construct an intelligent nanoplatform with synergistic therapeutic effect is still a great challenge. In this work, curcumin-loaded reduction-responsive prodrug nanoparticles of new indocyanine green (Cur@IR820-ss-PEG) were developed for synergistic cancer chemo-photodynamic therapy. Cur@IR820-ss-PEG exhibit high drug loading content and special worm-like morphology, contributing to their efficient cellular uptake. Due to the presence of the disulfide bond between IR820 and PEG, Cur@IR820-ss-PEG display reduction responsive drug release behaviors. The efficient cellular uptake and reduction triggered drug release of Cur@IR820-ss-PEG lead to their enhanced in vitro cytotoxicity against 4T1cells as compared to the mixture of IR820 and curcumin (IR820/Cur) under laser irradiation. Besides, Cur@IR820-ss-PEG exhibit prolonged blood half-life time, better tumor accumulation and retention, enhanced tumor hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial cell growth factor (VEGF) suppression effect as compared to IR820/Cur. In vivo antitumor activity study, Cur@IR820-ss-PEG effectively inhibit the tumor angiogenesis, which potentiates the PDT efficacy and leads to the best in vivo antitumor effect of Cur@IR820-ss-PEG. This work provides a novel and relatively simple strategy for synergistic cancer chemo-photodynamic therapy.
Collapse
Affiliation(s)
- Hang Hu
- Second People's Hospital of Changzhou, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Defeng Xu
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China
| | - Qingbo Xu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Interventional Medical Center, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
| | - Yuxiang Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Jun Hong
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China,Corresponding author. Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jianhao Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China,Corresponding author.
| | - Xinye Ni
- Second People's Hospital of Changzhou, Nanjing Medical University, Changzhou, Jiangsu, China,Corresponding author.
| |
Collapse
|
29
|
Pola M, Kolarova H, Bajgar R. Generation of singlet oxygen by porphyrin and phthalocyanine derivatives regarding the oxygen level. JOURNAL OF MEDICAL SCIENCE 2022. [DOI: 10.20883/medical.e752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background. The principle of photodynamic effect is based on the combined action of photosensitiser, molecular oxygen and light, which produce various reactive oxygen species and are associated with significant cellular damage. Singlet oxygen is one of the most serious representatives, which is characterised by powerful oxidising properties. Moreover, concomitant hyperbaric oxygen treatment can support these effects. Therefore, the subject of our study was to compare the yields of singlet oxygen for four different photosensitizers in dependency on the oxygen concentration.
Material and methods. Four different photosensitizers 5,10,15,20-tetrakis(1-methyl-4-pyridinio)porphyrin tetra(p-toluenesulfonate), tetramethylthionine chloride, 5,10,15,20-tetrakis(4-sulfonatophenyl)porphyrin zinc(II) and zinc phthalocyanine disulfonate were investigated to determine the yield of singlet oxygen in PBS by Singlet Oxygen Sensor Green reagent under different partial pressures of oxygen (0.4 and 36 mg/l).
Results. There were no noticeable shifts in the excitation and emission fluorescence spectra regarding the oxygen concentration. Concerning the same molar concentration of photosensitizers the production of singlet oxygen was highest for 5,10,15,20-tetrakis(4-sulfonatophenyl)porphyrin zinc(II), where the rate of the fluorescence change was more than 3 times higher than that obtained for 5,10,15,20-tetrakis(1-methyl-4-pyridinio)porphyrin tetra(p-toluenesulfonate). On the other hand, zinc phthalocyanine disulfonate showed the lowest yield in singlet oxygen production.
Conclusions. Singlet oxygen production, within the range of oxygen concentrations achievable in tissues under normoxia or hyperoxia, does not depend on these concentrations. However, the singlet oxygen generation is significantly influenced by the type of photosensitizer, with the highest yield belonging to 5,10,15,20-tetrakis(4-sulfonatophenyl)porphyrin zinc(II).
Collapse
|
30
|
Jeon SI, Kim HJ, Lee JH, Ahn CH. Development of a Hypoxia-Sensitive Material Producing Fluorescence and Ultrasound Signals. Macromol Res 2022. [DOI: 10.1007/s13233-022-0100-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
31
|
Lei L, Huang D, Gao H, He B, Cao J, Peppas NA. Hydrogel-guided strategies to stimulate an effective immune response for vaccine-based cancer immunotherapy. SCIENCE ADVANCES 2022; 8:eadc8738. [PMID: 36427310 PMCID: PMC9699680 DOI: 10.1126/sciadv.adc8738] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 10/07/2022] [Indexed: 05/25/2023]
Abstract
Cancer vaccines have attracted widespread interest in tumor therapy because of the potential to induce an effective antitumor immune response. However, many challenges including weak immunogenicity, off-target effects, and immunosuppressive microenvironments have prevented their broad clinical translation. To overcome these difficulties, effective delivery systems have been designed for cancer vaccines. As carriers in cancer vaccine delivery systems, hydrogels have gained substantial attention because they can encapsulate a variety of antigens/immunomodulators and protect them from degradation. This enables hydrogels to simultaneously reverse immunosuppression and stimulate the immune response. Meanwhile, the controlled release properties of hydrogels allow for precise temporal and spatial release of loads in situ to further enhance the immune response of cancer vaccines. Therefore, this review summarizes the classification of cancer vaccines, highlights the strategies of hydrogel-based cancer vaccines, and provides some insights into the future development of hydrogel-based cancer vaccines.
Collapse
Affiliation(s)
- Lei Lei
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China
| | - Dennis Huang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX 78712, USA
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| | - Jun Cao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| | - Nicholas A. Peppas
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
- Departments of Pediatrics, Surgery, and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
32
|
Zhang L, Oudeng G, Wen F, Liao G. Recent advances in near-infrared-II hollow nanoplatforms for photothermal-based cancer treatment. Biomater Res 2022; 26:61. [PMID: 36348441 PMCID: PMC9641873 DOI: 10.1186/s40824-022-00308-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/16/2022] [Indexed: 11/10/2022] Open
Abstract
Near-infrared-II (NIR-II, 1000–1700 nm) light-triggered photothermal therapy (PTT) has been regarded as a promising candidate for cancer treatment, but PTT alone often fails to achieve satisfactory curative outcomes. Hollow nanoplatforms prove to be attractive in the biomedical field owing to the merits including good biocompatibility, intrinsic physical-chemical nature and unique hollow structures, etc. On one hand, hollow nanoplatforms themselves can be NIR-II photothermal agents (PTAs), the cavities of which are able to carry diverse therapeutic units to realize multi-modal therapies. On the other hand, NIR-II PTAs are capable of decorating on the surface to combine with the functions of components encapsulated inside the hollow nanoplatforms for synergistic cancer treatment. Notably, PTAs generally can serve as good photoacoustic imaging (PAI) contrast agents (CAs), which means such kind of hollow nanoplatforms are also expected to be multifunctional all-in-one nanotheranostics. In this review, the recent advances of NIR-II hollow nanoplatforms for single-modal PTT, dual-modal PTT/photodynamic therapy (PDT), PTT/chemotherapy, PTT/catalytic therapy and PTT/gas therapy as well as multi-modal PTT/chemodynamic therapy (CDT)/chemotherapy, PTT/chemo/gene therapy and PTT/PDT/CDT/starvation therapy (ST)/immunotherapy are summarized for the first time. Before these, the typical synthetic strategies for hollow structures are presented, and lastly, potential challenges and perspectives related to these novel paradigms for future research and clinical translation are discussed.
Collapse
|
33
|
Pewklang T, Chansaenpak K, Bakar SN, Lai RY, Kue CS, Kamkaew A. Aza-BODIPY based carbonic anhydrase IX: Strategy to overcome hypoxia limitation in photodynamic therapy. Front Chem 2022; 10:1015883. [PMID: 36405312 PMCID: PMC9666899 DOI: 10.3389/fchem.2022.1015883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/19/2022] [Indexed: 11/28/2022] Open
Abstract
Hypoxia caused by photodynamic therapy (PDT) is a major hurdle to cancer treatment since it can promote recurrence and progression by activating angiogenic factors, lowering therapeutic efficacy dramatically. In this work, AZB-I-CAIX2 was developed as a carbonic anhydrase IX (CAIX)-targeting NIR photosensitizer that can overcome the challenge by utilizing a combination of CAIX knockdown and PDT. AZB-I-CAIX2 showed a specific affinity to CAIX-expressed cancer cells and enhanced photocytotoxicity compared to AZB-I-control (the molecule without acetazolamide). Moreover, selective detection and effective cell cytotoxicity of AZB-I-CAIX2 by PDT in hypoxic CAIX-expressed murine cancer cells were achieved. Essentially, AZB-I-CAIX2 could minimize tumor size in the tumor-bearing mice compared to that in the control groups. The results suggested that AZB-I-CAIX2 can improve therapeutic efficiency by preventing PDT-induced hypoxia through CAIX inhibition.
Collapse
Affiliation(s)
- Thitima Pewklang
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Kantapat Chansaenpak
- National Nanotechnology Center, National Science and Technology Development Agency, Thailand Science Park, Pathum Thani, Thailand
| | - Siti Nursyahirah Bakar
- Faculty of Health and Life Sciences, Management and Science University, Shah Alam, Selangor, Malaysia
| | - Rung-Yi Lai
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Chin Siang Kue
- Faculty of Health and Life Sciences, Management and Science University, Shah Alam, Selangor, Malaysia,*Correspondence: Anyanee Kamkaew, ; Chin Siang Kue,
| | - Anyanee Kamkaew
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand,*Correspondence: Anyanee Kamkaew, ; Chin Siang Kue,
| |
Collapse
|
34
|
Yang DC, Wen LF, Du L, Luo CM, Lu ZY, Liu JY, Lin Z. A Hypoxia-Activated Prodrug Conjugated with a BODIPY-Based Photothermal Agent for Imaging-Guided Chemo-Photothermal Combination Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:40546-40558. [PMID: 36059107 DOI: 10.1021/acsami.2c09071] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Hypoxia-activated prodrugs (HAPs) have drawn increasing attention for improving the antitumor effects while minimizing side effects. However, the heterogeneous distribution of the hypoxic region in tumors severely impedes the curative effect of HAPs. Additionally, most HAPs are not amenable to optical imaging, and it is difficult to precisely trace them in tissues. Herein, we carefully designed and synthesized a multifunctional therapeutic BAC prodrug by connecting the chemotherapeutic drug camptothecin (CPT) and the fluorescent photothermal agent boron dipyrromethene (BODIPY) via hypoxia-responsive azobenzene linkers. To enhance the solubility and tumor accumulation, the prepared BAC was further encapsulated into a human serum albumin (HSA)-based drug delivery system to form HSA@BAC nanoparticles. Since the CPT was caged by a BODIPY-based molecule at the active site, the BAC exhibited excellent biosafety. Importantly, the activated CPT could be quickly released from BAC and could perform chemotherapy in hypoxic cancer cells, which was ascribed to the cleavage of the azobenzene linker by overexpressed azoreductase. After irradiation with a 730 nm laser, HSA@BAC can efficiently generate hyperthermia to achieve irreversible cancer cell death by oxygen-independent photothermal therapy. Under fluorescence imaging-guided local irradiation, both in vitro and in vivo studies demonstrated that HSA@BAC exhibited superior antitumor effects with minimal side effects.
Collapse
Affiliation(s)
- De-Chao Yang
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Lin-Feng Wen
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Liyang Du
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Cheng-Miao Luo
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Zi-Yao Lu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Jian-Yong Liu
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
- Key Laboratory of Molecule Synthesis and Function Discovery, Fujian Province University, College of Chemistry, Fuzhou University, Fuzhou 350108, China
- State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Zhonghui Lin
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| |
Collapse
|
35
|
Chen W, Du W, Zhang H, Cheng L, Song L, Ma X, Hu Y, Wang J. Hemin-loaded black phosphorus-based nanosystem for enhanced photodynamic therapy and a synergistic photothermally/photodynamically activated inflammatory immune response. BIOMATERIALS ADVANCES 2022; 140:213091. [PMID: 36041322 DOI: 10.1016/j.bioadv.2022.213091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 08/12/2022] [Accepted: 08/20/2022] [Indexed: 06/15/2023]
Abstract
The biocompatible nanosystem integrating hemin into black phosphorus nanosheets was ingeniously constructed through the easy modified strategy. Taking advantage of the enhanced permeability and retention (EPR) effect, the designed nanosystem could accumulate into the tumor location, leading to attractive cytotoxicity through the enhanced photodynamic therapy (PDT) ascribing to the catalytic oxygen supply and GSH depletion of hemin. Simultaneously, combining PDT and photothermal therapy (PTT) showed an apparent promotion in anti-tumor effect. Moreover, inflammatory response and immune activation amplified anti-tumor effect, which could compensate limitations of exogenous therapy (i.e., limited tissue depth and intensity-dependent curation effect) and potentiate the efficiency of the endogenous immune-activating behavior. Especially, the designed nanosystem degraded followed by being metabolized in the blood circulation. By and large, this constructed nanosystem provides the new insight into designing biocompatible nanomaterials and paves the ideal way for anti-tumor therapy. STATEMENT OF SIGNIFICANCE: Biocompatible nanomaterials-based synergistic tumor therapy offers the potential application prospect. Taking advantage of degradable black phosphorus, the nanosystem integrating hemin into black phosphorus for the enhanced photodynamic therapy and synergistic photothermal-photodynamic activating inflammation-immune response was developed and the results demonstrate that tumor growth was inhibited followed by activating inflammatory factors and leading to satisfactory immune response.
Collapse
Affiliation(s)
- Weijian Chen
- School of Chemistry and Materials Science, University of Science and Technology of China, Jinzhai Road 96, Hefei, Anhui, PR China; Department of thyroid and breast Surgery, The First Affiliated Hospital of USTC, Tian'ehu Road 1, Hefei, Anhui, PR China; State Key Laboratory of Fire Science, University of Science and Technology of China, Huangshan Road 443, Hefei, Anhui, PR China
| | - Wenxiang Du
- School of Chemistry and Materials Science, University of Science and Technology of China, Jinzhai Road 96, Hefei, Anhui, PR China; Department of thyroid and breast Surgery, The First Affiliated Hospital of USTC, Tian'ehu Road 1, Hefei, Anhui, PR China; State Key Laboratory of Fire Science, University of Science and Technology of China, Huangshan Road 443, Hefei, Anhui, PR China
| | - Hongjie Zhang
- School of Chemistry and Materials Science, University of Science and Technology of China, Jinzhai Road 96, Hefei, Anhui, PR China; Department of thyroid and breast Surgery, The First Affiliated Hospital of USTC, Tian'ehu Road 1, Hefei, Anhui, PR China; State Key Laboratory of Fire Science, University of Science and Technology of China, Huangshan Road 443, Hefei, Anhui, PR China
| | - Liang Cheng
- State Key Laboratory of Fire Science, University of Science and Technology of China, Huangshan Road 443, Hefei, Anhui, PR China
| | - Lei Song
- State Key Laboratory of Fire Science, University of Science and Technology of China, Huangshan Road 443, Hefei, Anhui, PR China
| | - Xiaopeng Ma
- Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, Anhui, PR China; Department of thyroid and breast Surgery, The First Affiliated Hospital of USTC, Tian'ehu Road 1, Hefei, Anhui, PR China
| | - Yuan Hu
- School of Chemistry and Materials Science, University of Science and Technology of China, Jinzhai Road 96, Hefei, Anhui, PR China; State Key Laboratory of Fire Science, University of Science and Technology of China, Huangshan Road 443, Hefei, Anhui, PR China.
| | - Jing Wang
- Department of thyroid and breast Surgery, The First Affiliated Hospital of USTC, Tian'ehu Road 1, Hefei, Anhui, PR China; State Key Laboratory of Fire Science, University of Science and Technology of China, Huangshan Road 443, Hefei, Anhui, PR China.
| |
Collapse
|
36
|
Carrier free nanomedicine for synergistic cancer therapy by initiating apoptosis and paraptosis. J Colloid Interface Sci 2022; 622:298-308. [DOI: 10.1016/j.jcis.2022.04.090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 02/06/2023]
|
37
|
Du Y, Han J, Jin F, Du Y. Recent Strategies to Address Hypoxic Tumor Environments in Photodynamic Therapy. Pharmaceutics 2022; 14:pharmaceutics14091763. [PMID: 36145513 PMCID: PMC9505114 DOI: 10.3390/pharmaceutics14091763] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/15/2022] [Accepted: 08/22/2022] [Indexed: 12/02/2022] Open
Abstract
Photodynamic therapy (PDT) has become a promising method of cancer treatment due to its unique properties, such as noninvasiveness and low toxicity. The efficacy of PDT is, however, significantly reduced by the hypoxia tumor environments, because PDT involves the generation of reactive oxygen species (ROS), which requires the great consumption of oxygen. Moreover, the consumption of oxygen caused by PDT would further exacerbate the hypoxia condition, which leads to angiogenesis, invasion of tumors to other parts, and metastasis. Therefore, many research studies have been conducted to design nanoplatforms that can alleviate tumor hypoxia and enhance PDT. Herein, the recent progress on strategies for overcoming tumor hypoxia is reviewed, including the direct transport of oxygen to the tumor site by O2 carriers, the in situ generation of oxygen by decomposition of oxygen-containing compounds, reduced O2 consumption, as well as the regulation of tumor microenvironments. Limitations and future perspectives of these technologies to improve PDT are also discussed.
Collapse
|
38
|
Thomas CD, Lupu M, Poyer F, Maillard P, Mispelter J. Increased PDT Efficacy When Associated with Nitroglycerin: A Study on Retinoblastoma Xenografted on Mice. Pharmaceuticals (Basel) 2022; 15:ph15080985. [PMID: 36015132 PMCID: PMC9415823 DOI: 10.3390/ph15080985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/02/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Purposes: The aim of the study was to assess the efficacy of a treatment protocol that combines photodynamic therapy (PDT) and nitroglycerin (NG) on human retinoblastoma tumors xenografted on mice. We aimed to increase the PDT efficiency (in our least treatment-responsive retinoblastoma line) with better PS delivery to the tumor generated by NG, which is known to dilate vessels and enhance the permeability and retention of macromolecules in solid tumors. Methods: In vivo follow-up of the therapeutic effects was performed by sodium MRI, which directly monitors variations in sodium concentrations non-invasively and can be used to track the tumor response to therapy. NG ointment was applied one hour before PDT. The PDT protocol involves double-tumor targeting, i.e., cellular and vascular. The first PS dose was injected followed by a second one, separated by a 3 h interval. The timelapse allowed the PS molecules to penetrate tumor cells. Ten minutes after the second dose, the PS was red-light-activated. Results: In this study, we observed that the PDT effect was enhanced by applying nitroglycerin ointment to the tumor-bearing animal’s skin. PDT initiates the bystander effect on retinoblastomas, and NG increases this effect by increasing the intratumoral concentration of PS, which induces a higher production of ROS in the illuminated region and thus increases the propagation of the cell death signal deeper into the tumor (bystander effect).
Collapse
Affiliation(s)
- Carole D. Thomas
- Institut Curie, Bât. 112, Centre Universitaire, 91405 Orsay, France
- U1288 INSERM, LITO Laboratoire d’Imagerie Translationnelle en Oncologie, Bât. 101B, Centre Universitaire, 91405 Orsay, France
- Université Paris-Sud, 91405 Orsay, France
- Correspondence: ; Tel.: +33-(0)1-69-86-71-97
| | - Mihaela Lupu
- Institut Curie, Bât. 112, Centre Universitaire, 91405 Orsay, France
- Université Paris-Sud, 91405 Orsay, France
- U1196 INSERM, Bât. 112, Centre Universitaire, 91405 Orsay, France
- UMR 9187 CNRS, Bât. 112, Centre Universitaire, 91405 Orsay, France
| | - Florent Poyer
- Institut Curie, Bât. 112, Centre Universitaire, 91405 Orsay, France
- Université Paris-Sud, 91405 Orsay, France
- U1196 INSERM, Bât. 112, Centre Universitaire, 91405 Orsay, France
- UMR 9187 CNRS, Bât. 112, Centre Universitaire, 91405 Orsay, France
| | - Philippe Maillard
- Institut Curie, Bât. 112, Centre Universitaire, 91405 Orsay, France
- Université Paris-Sud, 91405 Orsay, France
- U1196 INSERM, Bât. 112, Centre Universitaire, 91405 Orsay, France
- UMR 9187 CNRS, Bât. 112, Centre Universitaire, 91405 Orsay, France
| | - Joël Mispelter
- Institut Curie, Bât. 112, Centre Universitaire, 91405 Orsay, France
- Université Paris-Sud, 91405 Orsay, France
- U1196 INSERM, Bât. 112, Centre Universitaire, 91405 Orsay, France
- UMR 9187 CNRS, Bât. 112, Centre Universitaire, 91405 Orsay, France
| |
Collapse
|
39
|
Sarbadhikary P, George BP, Abrahamse H. Potential Application of Photosensitizers With High-Z Elements for Synergic Cancer Therapy. Front Pharmacol 2022; 13:921729. [PMID: 35837287 PMCID: PMC9274123 DOI: 10.3389/fphar.2022.921729] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/17/2022] [Indexed: 01/10/2023] Open
Abstract
The presence of heavy elements in photosensitizers (PS) strongly influences their electronic and photophysical properties, and hence, conjugation of PS with a suitable element is regarded as a potential strategy to improve their photodynamic properties. Moreover, PS conjugated to metal ion or metal complex and heavy atoms such as halogen have attracted considerable attention as promising agents for multimodal or synergistic cancer therapy. These tetrapyrrole compounds depending on the type and nature of the inorganic elements have been explored for photodynamic therapy (PDT), chemotherapy, X-ray photon activation therapy (PAT), and radiotherapy. Particularly, the combination of metal-based PS and X-ray irradiation has been investigated as a promising novel approach for treating deep-seated tumors, which in the case of PDT is a major limitation due to low light penetration in tissue. This review will summarize the present status of evidence on the effect of insertion of metal or halogen on the photophysical properties of PS and the effectiveness of various metal and halogenated PS investigated for PDT, chemotherapy, and PAT as mono and/or combination therapy.
Collapse
|
40
|
Chen K, Li H, Zhou A, Zhou X, Xu Y, Ge H, Ning X. Cell Membrane Camouflaged Metal Oxide-Black Phosphorus Biomimetic Nanocomplex Enhances Photo-chemo-dynamic Ferroptosis. ACS APPLIED MATERIALS & INTERFACES 2022; 14:26557-26570. [PMID: 35658416 DOI: 10.1021/acsami.2c08413] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Despite the availability of various treatment options, the inherent complexity of tumors significantly impairs therapeutic efficacy. Recently, combination treatments exhibited great anticancer potential due to low cross-resistance and good therapeutic additivity. Herein, a photoactive metal oxide-black phosphorus biomimetic nanocomplex (photophage) is developed for improving the antitumor combination of ferroptosis and photodynamic therapy (PDT). The photophage is composed of M1 macrophage membrane camouflaged MnO2 and Fe3O4 nanoparticles anchored black phosphorus nanosheets (BPNs), which together trigger a synergistic antitumor action. Fe3O4 acts as an iron source to activate Fenton-reaction-dependent ferroptosis, which can be further strengthened by BPN-mediated PDT. Besides the original antitumor effects, PDT also generates reactive oxygen species to enhance lipid peroxidation and glutathione depletion, which in turn reinforce ferroptosis and PDT efficacy. Importantly, MnO2 can in situ generate oxygen to relieve tumor hypoxia and consequently leverage cell behaviors to improve therapeutic responses. Particularly, M1 macrophage membrane modification endows the photophage with good tumor targeting capability and tumor penetration, which promote synergistic ferroptosis and PDT to destroy tumors while reducing systemic side effects, resulting in the prolonged survival of tumor-bearing mice. Therefore, we present a biomimetic nanoplatform for overcoming tumor barriers and advancing tumor-targeted treatment.
Collapse
Affiliation(s)
- Kerong Chen
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, China
| | - Huipeng Li
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, China
- Center for Health Science and Engineering, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300131, China
| | - Anwei Zhou
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, School of Physics, Nanjing University, Nanjing 210093, China
| | - Xinyuan Zhou
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, China
| | - Yurui Xu
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, China
| | - Haixiong Ge
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, China
| | - Xinghai Ning
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, China
| |
Collapse
|
41
|
Photodynamic Therapy-Adjunctive Therapy in the Treatment of Prostate Cancer. Diagnostics (Basel) 2022; 12:diagnostics12051113. [PMID: 35626269 PMCID: PMC9139878 DOI: 10.3390/diagnostics12051113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 12/25/2022] Open
Abstract
The alarming increase in the number of advanced-stage prostate cancer cases with poor prognosis has led to a search for innovative methods of treatment. In response to the need for implementation of new and innovative methods of cancer tissue therapy, we studied photodynamic action in excised prostate tissue in vitro as a model for photodynamic therapy. To ascertain the effects of photodynamic action in prostate tissue, Rose Bengal (0.01 to 0.05 mM) was used as a photosensitizer in the presence of oxygen and light to generate singlet oxygen in tissues in vitro. Five preset concentrations of Rose Bengal were chosen and injected into prostate tissue samples (60 samples with 12 replications for each RB concentration) that were subsequently exposed to 532 nm light. The effects of irradiation of the Rose Bengal infused tissue samples were determined by histopathological analysis. Histopathological examination of prostate samples subjected to photodynamic action revealed numerous changes in the morphology of the neoplastic cells and the surrounding tissues. We conclude that the morphological changes observed in the prostate cancer tissues were a result of the photogeneration of cytotoxic singlet oxygen. The tissue damage observed post photodynamic action offers an incentive for continued in vitro investigations and future in vivo clinical trials.
Collapse
|
42
|
Pucelik B, Sułek A, Borkowski M, Barzowska A, Kobielusz M, Dąbrowski JM. Synthesis and Characterization of Size- and Charge-Tunable Silver Nanoparticles for Selective Anticancer and Antibacterial Treatment. ACS APPLIED MATERIALS & INTERFACES 2022; 14:14981-14996. [PMID: 35344328 PMCID: PMC8990520 DOI: 10.1021/acsami.2c01100] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Advances in the research of nanoparticles (NPs) with controlled charge and size are driven by their potential application in the development of novel technologies and innovative therapeutics. This work reports the synthesis, characterization, and comprehensive biological evaluation of AgNPs functionalized by N,N,N-trimethyl-(11-mercaptoundecyl) ammonium chloride (TMA) and trisodium citrate (TSC). The prepared AgNPs were well characterized in terms of their morphological, spectroscopic and functional properties and biological activities. The implementation of several complementary techniques allowed not only the estimation of the average particle size (from 3 to 40 nm depending on the synthesis procedure used) but also the confirmation of the crystalline nature of the NPs and their round shape. To prove the usefulness of these materials in biological systems, cellular uptake and cytotoxicity in microbial and mammalian cells were determined. Positively charged 10 nm Ag@TMA2 revealed antimicrobial activity against Gram-negative bacteria with a minimum inhibitory concentration (MIC) value of 0.17 μg/mL and complete eradication of Escherichia coli (7 logs) for Ag@TMA2 at a concentration of 0.50 μg/mL, whereas negatively charged 10 nm Ag@TSC1 was effective against Gram-positive bacteria (MIC = 0.05 μg/mL), leading to inactivation of Staphylococcus aureus at relatively low concentrations. In addition, the largest 40 nm Ag@TSC2 was shown to exhibit pronounced anticancer activity against murine colon carcinoma (CT26) and murine mammary gland carcinoma (4T1) cells cultured as 2D and 3D tumor models and reduced toxicity against human HaCaT keratinocytes. Among the possible mechanisms of AgNPs are their ability to generate reactive oxygen species, which was further evaluated in vitro and correlates well with cellular accumulation and overall activity of AgNPs. Furthermore, we confirmed the anticancer efficacy of the most potent Ag@TSC2 in hiPSC-derived colonic organoids and demonstrated that the NPs are biocompatible and applicable in vivo. A pilot study in BALB/c mice evidenced that the treatment with Ag@TSC2 resulted in temporary (>60 days) remission of CT26 tumors.
Collapse
Affiliation(s)
- Barbara Pucelik
- Małopolska
Centre of Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Adam Sułek
- Faculty
of Chemistry, Jagiellonian University, 30-387 Kraków, Poland
| | - Mariusz Borkowski
- Jerzy
Haber Institute of Catalysis and Surface Chemistry Polish Academy
of Sciences, 30-239 Kraków, Poland
| | - Agata Barzowska
- Małopolska
Centre of Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Marcin Kobielusz
- Faculty
of Chemistry, Jagiellonian University, 30-387 Kraków, Poland
| | | |
Collapse
|
43
|
Jiang M, Liu Y, Dong Y, Wang K, Yuan Y. Bioorthogonal chemistry and illumination controlled programmed size-changeable nanomedicine for synergistic photodynamic and hypoxia-activated therapy. Biomaterials 2022; 284:121480. [DOI: 10.1016/j.biomaterials.2022.121480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 03/16/2022] [Accepted: 03/19/2022] [Indexed: 11/25/2022]
|
44
|
Merabti A, Roger M, Nguyen C, Nocentini A, Gerbier P, Richeter S, Gary‐Bobo M, Supuran CT, Clément S, Winum J. Carbonic Anhydrase Inhibitors Featuring a Porphyrin Scaffold: Synthesis, Optical and Biological Properties. European J Org Chem 2022. [DOI: 10.1002/ejoc.202101538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Amina Merabti
- IBMM Univ Montpellier CNRS ENSCM Montpellier France
- ICGM Univ Montpellier CNRS ENSCM Montpellier France
| | - Maxime Roger
- ICGM Univ Montpellier CNRS ENSCM Montpellier France
| | | | - Alessio Nocentini
- Neurofarba Department Sezione Di Chimica Farmaceutica E Nutraceutica Università Degli Studi Di Firenze Via U. Schiff 6 50019, Sesto Fiorentino Firenze Italy
| | | | | | | | - Claudiu T. Supuran
- Neurofarba Department Sezione Di Chimica Farmaceutica E Nutraceutica Università Degli Studi Di Firenze Via U. Schiff 6 50019, Sesto Fiorentino Firenze Italy
| | | | | |
Collapse
|
45
|
Mazuryk O, Janczy-Cempa E, Łagosz J, Rutkowska-Zbik D, Machnicka A, Krasowska A, Pietrzyk P, Stochel G, Brindell M. Relevance of the electron transfer pathway in photodynamic activity of Ru(II) polypyridyl complexes containing 4,7-diphenyl-1,10-phenanthroline ligands under normoxic and hypoxic conditions. Dalton Trans 2022; 51:1888-1900. [PMID: 35018930 DOI: 10.1039/d1dt02908h] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The purpose of this study was to investigate the correlation between the spectroscopic and photophysical properties of Ru(II) polypyridyl complexes and their photodynamic activity in vitro. A series of Ru(II) polypyridyl complexes with 4,7-diphenyl-1,10-phenanthroline (dip) and 2,3-bis(2-pyridyl)quinoxaline (dpq) and its derivatives were synthesized and characterized regarding their photophysical, biological, and photodynamic properties. The complexes were evaluated not only in the context of 1O2 generation but also regarding other types of reactive oxygen species (ROS) to assess the possibility of Ru(II) complexes to induce phototoxicity via various ROS using fluorescence and EPR spectroscopy. The compounds were found to be moderately cytotoxic with IC50 values ranging from 1 to 35 μM and retained their cytotoxic activity under hypoxic conditions. The unraveled phototoxic activity is based mainly on the generation of H2O2 and 1O2, highlighting the importance of electron-transfer processes in the observed photodynamic activity of Ru polypyridyl complexes. A combination of photodynamic activity with cytotoxicity under decreased dioxygen concentrations may help overcome the current photodynamic therapy (PDT) limitation. The findings highlight the need for broadening the scope of tested Ru-based photosensitizers.
Collapse
Affiliation(s)
- Olga Mazuryk
- Department of Inorganic Chemistry, Faculty of Chemistry, Jagiellonian University in Kraków, Gronostajowa 2, 30-387 Krakow, Poland.
| | - Ewelina Janczy-Cempa
- Department of Inorganic Chemistry, Faculty of Chemistry, Jagiellonian University in Kraków, Gronostajowa 2, 30-387 Krakow, Poland.
| | - Justyna Łagosz
- Department of Inorganic Chemistry, Faculty of Chemistry, Jagiellonian University in Kraków, Gronostajowa 2, 30-387 Krakow, Poland.
| | - Dorota Rutkowska-Zbik
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek 8, 30-239 Krakow, Poland
| | - Agata Machnicka
- Department of Inorganic Chemistry, Faculty of Chemistry, Jagiellonian University in Kraków, Gronostajowa 2, 30-387 Krakow, Poland.
| | - Aneta Krasowska
- Department of Inorganic Chemistry, Faculty of Chemistry, Jagiellonian University in Kraków, Gronostajowa 2, 30-387 Krakow, Poland.
| | - Piotr Pietrzyk
- Department of Inorganic Chemistry, Faculty of Chemistry, Jagiellonian University in Kraków, Gronostajowa 2, 30-387 Krakow, Poland.
| | - Grażyna Stochel
- Department of Inorganic Chemistry, Faculty of Chemistry, Jagiellonian University in Kraków, Gronostajowa 2, 30-387 Krakow, Poland.
| | - Małgorzata Brindell
- Department of Inorganic Chemistry, Faculty of Chemistry, Jagiellonian University in Kraków, Gronostajowa 2, 30-387 Krakow, Poland.
| |
Collapse
|
46
|
Pucelik B, Dąbrowski JM. Photodynamic inactivation (PDI) as a promising alternative to current pharmaceuticals for the treatment of resistant microorganisms. ADVANCES IN INORGANIC CHEMISTRY 2022; 79:65-103. [PMID: 35095189 PMCID: PMC8787646 DOI: 10.1016/bs.adioch.2021.12.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Although the whole world is currently observing the global battle against COVID-19, it should not be underestimated that in the next 30 years, approximately 10 million people per year could be exposed to infections caused by multi-drug resistant bacteria. As new antibiotics come under pressure from unpredictable resistance patterns and relegation to last-line therapy, immediate action is needed to establish a radically different approach to countering resistant microorganisms. Among the most widely explored alternative methods for combating bacterial infections are metal complexes and nanoparticles, often in combination with light, but strategies using monoclonal antibodies and bacteriophages are increasingly gaining acceptance. Photodynamic inactivation (PDI) uses light and a dye termed a photosensitizer (PS) in the presence of oxygen to generate reactive oxygen species (ROS) in the field of illumination that eventually kill microorganisms. Over the past few years, hundreds of photomaterials have been investigated, seeking ideal strategies based either on single molecules (e.g., tetrapyrroles, metal complexes) or in combination with various delivery systems. The present work describes some of the most recent advances of PDI, focusing on the design of suitable photosensitizers, their formulations, and their potential to inactivate bacteria, viruses, and fungi. Particular attention is focused on the compounds and materials developed in our laboratories that are capable of killing in the exponential growth phase (up to seven logarithmic units) of bacteria without loss of efficacy or resistance, while being completely safe for human cells. Prospectively, PDI using these photomaterials could potentially cure infected wounds and oral infections caused by various multidrug-resistant bacteria. It is also possible to treat the surfaces of medical equipment with the materials described, in order to disinfect them with light, and reduce the risk of nosocomial infections.
Collapse
Affiliation(s)
- Barbara Pucelik
- Faculty of Chemistry, Jagiellonian University, Kraków, Poland
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Janusz M Dąbrowski
- Faculty of Chemistry, Jagiellonian University, Kraków, Poland
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
47
|
Recent Advances in Strategies for Addressing Hypoxia in Tumor Photodynamic Therapy. Biomolecules 2022; 12:biom12010081. [PMID: 35053229 PMCID: PMC8774200 DOI: 10.3390/biom12010081] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/20/2021] [Accepted: 12/29/2021] [Indexed: 01/10/2023] Open
Abstract
Photodynamic therapy (PDT) is a treatment modality that uses light to target tumors and minimize damage to normal tissues. It offers advantages including high spatiotemporal selectivity, low side effects, and maximal preservation of tissue functions. However, the PDT efficiency is severely impeded by the hypoxic feature of tumors. Moreover, hypoxia may promote tumor metastasis and tumor resistance to multiple therapies. Therefore, addressing tumor hypoxia to improve PDT efficacy has been the focus of antitumor treatment, and research on this theme is continuously emerging. In this review, we summarize state-of-the-art advances in strategies for overcoming hypoxia in tumor PDTs, categorizing them into oxygen-independent phototherapy, oxygen-economizing PDT, and oxygen-supplementing PDT. Moreover, we highlight strategies possessing intriguing advantages such as exceedingly high PDT efficiency and high novelty, analyze the strengths and shortcomings of different methods, and envision the opportunities and challenges for future research.
Collapse
|
48
|
Managing GSH elevation and hypoxia to overcome resistance of cancer therapies using functionalized nanocarriers. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2021.103022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
49
|
Wen J, Luo Y, Gao H, Zhang L, Wang X, Huang J, Shang T, Zhou D, Wang D, Wang Z, Li P, Wang Z. Mitochondria-targeted nanoplatforms for enhanced photodynamic therapy against hypoxia tumor. J Nanobiotechnology 2021; 19:440. [PMID: 34930284 PMCID: PMC8686264 DOI: 10.1186/s12951-021-01196-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022] Open
Abstract
Background Photodynamic therapy (PDT) is a promising therapeutic modality that can convert oxygen into cytotoxic reactive oxygen species (ROS) via photosensitizers to halt tumor growth. However, hypoxia and the unsatisfactory accumulation of photosensitizers in tumors severely diminish the therapeutic effect of PDT. In this study, a multistage nanoplatform is demonstrated to overcome these limitations by encapsulating photosensitizer IR780 and oxygen regulator 3-bromopyruvate (3BP) in poly (lactic-co-glycolic acid) (PLGA) nanocarriers. Results The as-synthesized nanoplatforms penetrated deeply into the interior region of tumors and preferentially remained in mitochondria due to the intrinsic characteristics of IR780. Meanwhile, 3BP could efficiently suppress oxygen consumption of tumor cells by inhibiting mitochondrial respiratory chain to further improve the generation of ROS. Furthermore, 3BP could abolish the excessive glycolytic capacity of tumor cells and lead to the collapse of ATP production, rendering tumor cells more susceptible to PDT. Successful tumor inhibition in animal models confirmed the therapeutic precision and efficiency. In addition, these nanoplatforms could act as fluorescence (FL) and photoacoustic (PA) imaging contrast agents, effectuating imaging-guided cancer treatment. Conclusions This study provides an ideal strategy for cancer therapy by concurrent oxygen consumption reduction, oxygen-augmented PDT, energy supply reduction, mitochondria-targeted/deep-penetrated nanoplatforms and PA/FL dual-modal imaging guidance/monitoring. It is expected that such strategy will provide a promising alternative to maximize the performance of PDT in preclinical/clinical cancer treatment. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-01196-6.
Collapse
Affiliation(s)
- Jiexin Wen
- Department of Ultrasound, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, People's Republic of China
| | - Yong Luo
- Department of Ultrasound, The First People's Hospital of Chongqing Liang Jiang New Area, Chongqing, 401121, People's Republic of China
| | - Hui Gao
- Department of Ultrasound, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, People's Republic of China
| | - Liang Zhang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Xiang Wang
- Department of Ultrasound, The Third Affiliated Hospital, Chongqing Medical University, Chongqing, 401120, People's Republic of China
| | - Ju Huang
- Department of Ultrasound, The Third Affiliated Hospital, Chongqing Medical University, Chongqing, 401120, People's Republic of China
| | - Tingting Shang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Di Zhou
- Department of Radiology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, 400042, People's Republic of China
| | - Dong Wang
- Department of Ultrasound, The First Affiliated Hospital, Chongqing Medical University, Chongqing, 400042, People's Republic of China
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Pan Li
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Zhaoxia Wang
- Department of Ultrasound, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, People's Republic of China.
| |
Collapse
|
50
|
Nkune NW, Abrahamse H. Nanoparticle-Based Drug Delivery Systems for Photodynamic Therapy of Metastatic Melanoma: A Review. Int J Mol Sci 2021; 22:12549. [PMID: 34830431 PMCID: PMC8620728 DOI: 10.3390/ijms222212549] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/11/2021] [Accepted: 10/26/2021] [Indexed: 12/12/2022] Open
Abstract
Metastatic melanoma (MM) is a skin malignancy arising from melanocytes, the incidence of which has been rising in recent years. It poses therapeutic challenges due to its resistance to chemotherapeutic drugs and radiation therapy. Photodynamic therapy (PDT) is an alternative non-invasive modality that requires a photosensitizer (PS), specific wavelength of light, and molecular oxygen. Several studies using conventional PSs have highlighted the need for improved PSs for PDT applications to achieve desired therapeutic outcomes. The incorporation of nanoparticles (NPs) and targeting moieties in PDT have appeared as a promising strategy to circumvent various drawbacks associated with non-specific toxicity, poor water solubility, and low bioavailability of the PSs at targeted tissues. Currently, most studies investigating new developments rely on two-dimensional (2-D) monocultures, which fail to accurately mimic tissue complexity. Therefore, three-dimensional (3-D) cell cultures are ideal models to resemble tumor tissue in terms of architectural and functional properties. This review examines various PS drugs, as well as passive and active targeted PS nanoparticle-mediated platforms for PDT treatment of MM on 2-D and 3-D models. The overall findings of this review concluded that very few PDT studies have been conducted within 3-D models using active PS nanoparticle-mediated platforms, and so require further investigation.
Collapse
Affiliation(s)
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa;
| |
Collapse
|