1
|
Feng Y, Wu Z, Liu H, Xu R. Combining photodynamic therapy and ATM inhibition using modified bovine serum albumin: A co-delivery nano platform for eliciting pyroptosis and apoptosis to fuel TNBC therapy. Int J Biol Macromol 2025; 307:142140. [PMID: 40112963 DOI: 10.1016/j.ijbiomac.2025.142140] [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: 12/17/2024] [Revised: 02/27/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
Triple-negative breast cancer (TNBC) presents a formidable challenge due to its poorest prognosis and limited array of treatment options available. Photodynamic therapy (PDT) has emerged as a potent therapeutic modality to generate intratumoral toxic reactive oxygen species (ROS) in combating refractory triple-negative breast cancer (TNBC). However, its therapeutic efficacy is compromised due to insufficient tumor accumulation and therapeutic resistance. Herein, an "all-in-one" tumor-therapeutic nanomedicine named HA@IR780@KU55933@BSA (HIKB) which integrated photosensitizer IR780 with ATM kinase inhibitor KU55933 was designed to facilitate drug delivery and target specific pathways involved in tumor PDT treatment resistance. Co-delivery of IR780 and KU55933 exacerbated intracellular ROS production, mitochondrial dysfunction and DNA damage to form a potent anti-TNBC therapeutic cyclical feedback loop and then induced pyroptosis and apoptosis of TNBC cells by activating the Caspase3/GSDME signaling pathway and regulating apoptosis-related protein expression, respectively. In vivo evaluations in the TNBC orthotopic xenograft mouse model demonstrated that the designed HIKB NPs could accumulate in tumor tissues and exert synergistic therapeutic effects. Altogether, this study described a self-assembling strategy for constructing an all-in-one nanomedicine that effectively integrates multiple therapeutic modalities to provide a comprehensive and systemic approach to tumor suppression.
Collapse
Affiliation(s)
- Yuao Feng
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zeliang Wu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hui Liu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China
| | - Rong Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China.
| |
Collapse
|
2
|
Vu NN, Ng KW, Jaitpal S, Negahdary M, Nguyen T, Kodam RS, Mabbott S. High Precision Automated Synthesis of Surface-Enhanced (Resonance) Raman Nanotags. ACS Sens 2025. [PMID: 40299759 DOI: 10.1021/acssensors.5c00068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
Batch-to-batch inconsistencies and time-intensive protocols remain significant challenges in conventional nanomaterial synthesis. Here, we present an automated system that precisely fabricates silica-coated gold nanostars (AuNS@SiO2) incorporating Raman reporters (4-MBA and IR-780) defined as nanotags, thereby enabling control over their morphological, optical, and spectroscopic properties. The resulting nanotags were comprehensively characterized through UV-vis spectrophotometry, transmission electron microscopy (TEM), surface enhanced (resonance) Raman spectroscopy (SE(R)RS) measurements, dynamic light scattering (DLS), and zeta potential analyzes. Compared to manual methods, our automated approach demonstrated higher reproducibility in both synthesis and the properties of the produced nanotags. Additionally, to underscore their potential in sensing applications, we functionalized the nanotags with streptavidin. By combining precise control over nanotags synthesis with robust characterization, this work establishes a new standard for the rapid and reliable production of advanced nanoplatforms for biomedical applications.
Collapse
Affiliation(s)
- Ngoc Nhu Vu
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzel Street, College Station, Texas 77843, United States
- Center for Remote Health Technologies & Systems, Texas A&M Engineering Experiment Station, 600 Discovery Drive, College Station, Texas 77840-3006, United States
| | - Ka Wai Ng
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzel Street, College Station, Texas 77843, United States
- Center for Remote Health Technologies & Systems, Texas A&M Engineering Experiment Station, 600 Discovery Drive, College Station, Texas 77840-3006, United States
| | - Siddhant Jaitpal
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzel Street, College Station, Texas 77843, United States
- Center for Remote Health Technologies & Systems, Texas A&M Engineering Experiment Station, 600 Discovery Drive, College Station, Texas 77840-3006, United States
| | - Masoud Negahdary
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzel Street, College Station, Texas 77843, United States
- Center for Remote Health Technologies & Systems, Texas A&M Engineering Experiment Station, 600 Discovery Drive, College Station, Texas 77840-3006, United States
| | - Tran Nguyen
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzel Street, College Station, Texas 77843, United States
- Center for Remote Health Technologies & Systems, Texas A&M Engineering Experiment Station, 600 Discovery Drive, College Station, Texas 77840-3006, United States
| | - Rohit Sai Kodam
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzel Street, College Station, Texas 77843, United States
- Center for Remote Health Technologies & Systems, Texas A&M Engineering Experiment Station, 600 Discovery Drive, College Station, Texas 77840-3006, United States
| | - Samuel Mabbott
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzel Street, College Station, Texas 77843, United States
- Center for Remote Health Technologies & Systems, Texas A&M Engineering Experiment Station, 600 Discovery Drive, College Station, Texas 77840-3006, United States
| |
Collapse
|
3
|
Wang L, Lin Y, Liu S, Jin C, Huang Y, Liang H, Sun X, Zhang K, Chen H, Zhang X, Wang F, Lin Z, Yan L, Chen M, Zhuge D, Chen Y. Treating and Protecting against Recurrent Vulvovaginal Candidiasis Using the Vaginal Epithelial Cell Membrane-Based Photoimmunotherapeutic Nanoplatform. ACS NANO 2025; 19:15537-15553. [PMID: 40235023 DOI: 10.1021/acsnano.4c16974] [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: 04/17/2025]
Abstract
Recurrent vulvovaginal candidiasis (RVVC) is an opportunistic infection predominantly caused by Candida albicans (C. albicans) and is particularly prevalent among individuals on immunosuppressants. Currently, there are no FDA-approved therapies for specifically controlling RVVC, mainly due to the need for therapeutics against RVVC that require both antifungal treatments to resolve active infections and strategies to prevent recurrence. This study introduces a biomimetic photoimmunotherapeutic nanoplatform consisting of an adjuvant-encapsulated polymeric core stabilized by a photosensitizer-loaded vaginal epithelial cell membrane coating to treat and protect against RVVC. With its cell membrane camouflaging, the nanoplatforms target and enhance adherence to the intravaginal site of C. albicans infection, allowing the nanoplatform to resist being flushed away by vaginal fluids. Upon subsequent near-infrared irradiation, the nanoplatform's targeted photothermal power effectively eliminates C. albicans while minimizing thermal damage to surrounding healthy tissue. Postphotothermal treatment, the generated C. albicans-based debris and candidalysin-captured nanoplatform (serving as a nanotoxoid), along with adjuvant, are processed by resident antigen-presenting cells to promote multiantigenic immunity. This response provides protection against secondary intravaginal C. albicans infection (RVVC model) and C. albicans-induced systemic infection even under immunosuppressive conditions (septicemia model). Notably, anti-C. albicans antibodies produced in the pretreated mice exhibit comparable affinity to clinically isolated C. albicans strains, indicating potential for clinical application. Overall, this study underscores the potential of the proposed photoimmunotherapeutic nanoplatform for the effective treatment and prevention of RVVC.
Collapse
Affiliation(s)
- Ledan Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yijing Lin
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Shuangshuang Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Chenjie Jin
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yunxuan Huang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Department of Pharmaceutics, School of Pharmaceutical Sciences of Wenzhou Medical University, Wenzhou 325035, China
| | - Hui Liang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo 315302, China
| | - Xueying Sun
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo 315302, China
| | - Kexin Zhang
- Research Center of Basic Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Hanxing Chen
- Research Center of Basic Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xufei Zhang
- Experimental Animal Center, Wenzhou Medical University, Wenzhou 325027, China
| | - Fang Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zhenkun Lin
- Research Center of Basic Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Linzhi Yan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Mengchun Chen
- Department of Pharmaceutics, School of Pharmaceutical Sciences of Wenzhou Medical University, Wenzhou 325035, China
- Department of Pharmacy, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo 315302, China
| | - Deli Zhuge
- Department of Pharmaceutics, School of Pharmaceutical Sciences of Wenzhou Medical University, Wenzhou 325035, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo 315302, China
| | - Yijie Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Department of Pharmaceutics, School of Pharmaceutical Sciences of Wenzhou Medical University, Wenzhou 325035, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo 315302, China
- Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
4
|
Tu L, Xing B, Ma S, Zou Z, Wang S, Feng J, Cheng M, Jin Y. A review on polysaccharide-based tumor targeted drug nanodelivery systems. Int J Biol Macromol 2025; 304:140820. [PMID: 39933669 DOI: 10.1016/j.ijbiomac.2025.140820] [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: 10/25/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/13/2025]
Abstract
The tumor-targeted drug delivery system (TTDNS) uses nanocarriers to transport chemotherapeutic agents to target tumor cells or tissues precisely. This innovative approach considerably increases the effective concentration of these drugs at the tumor site, thereby enhancing their therapeutic efficacy. Many chemotherapeutic agents face challenges, such as low bioavailability, high cytotoxicity, and inadequate drug resistance. To address these obstacles, TTDNS comprising natural polysaccharides have gained increasing popularity in the field of nanotechnology owing to their ability to improve safety, bioavailability, and biocompatibility while reducing toxicity. In addition, it enhances permeability and allows for controlled drug delivery and release. This review focuses on the sources of natural polysaccharides and their direct and indirect mechanisms of anti-tumor activity. We also explored the preparation of various polysaccharide-based nanocarriers, including nanoparticles, nanoemulsions, nanohydrogels, nanoliposomes, nanocapsules, nanomicelles, nanocrystals, and nanofibers. Furthermore, this review delves into the versatile applications of polysaccharide-based nanocarriers, elucidating their capabilities for in vivo targeting, controlled release, and responsiveness to endogenous and exogenous stimuli, such as pH, reactive oxygen species, glutathione, light, ultrasound, and magnetic fields. This sophisticated design substantially enhances the chemotherapeutic efficacy of the encapsulated drugs at tumor sites and provides a basis for preclinical and clinical research. However, the in vivo stability, drug loading, and permeability of these preparations into tumor tissues still need to be improved. Most of the currently developed biomarker-sensitive polysaccharide nanocarriers are still in the laboratory stage, more innovative delivery mechanisms and clinical studies are needed to develop commercial nanocarriers for medical use.
Collapse
Affiliation(s)
- Liangxing Tu
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Banghuai Xing
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Shufei Ma
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Zijian Zou
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Siying Wang
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Jianfang Feng
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China; Guangxi University of Chinese Medicine, Nanning 530200, PR China.
| | - Meng Cheng
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China.
| | - Yi Jin
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China.
| |
Collapse
|
5
|
Zhu HJ, Sun YY, Du Y, Zhou SY, Qu Y, Pang SY, Zhu S, Yang Y, Guo ZN. Albumin-seeking near-infrared-II probe evaluating blood-brain barrier disruption in stroke. J Nanobiotechnology 2024; 22:742. [PMID: 39609666 PMCID: PMC11606037 DOI: 10.1186/s12951-024-02973-9] [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: 07/17/2024] [Accepted: 11/03/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Blood-brain barrier (BBB) disruption after stroke is closely associated with brain tissue edema and neuronal injury, which requires accurate assessment. However, there is a lack of appropriate BBB imaging modality in vivo. As albumin in the blood could cross the damaged BBB into brain tissue after stroke, it serves as a biomarker for BBB disruption. Therefore, we aimed to develop an albumin-seeking near-infrared (NIR) probe to assess BBB disruption in stroke. RESULTS We proposed a chemoselective strategy for seeking albumin with NIR dyes and identified an optimal probe to evaluate BBB disruption in stroke. The probe combined a NIR fluorescent dye with inherent albumin-targeting moieties and exhibited high affinity and selectivity for binding to albumin. Using a mouse stroke model, the probe displayed a high-resolution visualization of the location and extent of BBB disruption in vivo and correlated well with BBB leakage measured by Evans blue ex vivo. A dual-channel NIR-II imaging was successfully used to simultaneously assess BBB disruption and cerebral perfusion after stroke. Furthermore, we applied this method to dynamically evaluate the BBB disruption process and reperfusion of thrombolytic therapy in a stroke model in real time, which showed excellent application value. CONCLUSIONS We developed an albumin-seeking NIR probe that accurately evaluated BBB disruption in a safe, non-invasive and real-time manner in various stroke models, and has a great potential guiding stroke treatment in a real-time manner.
Collapse
Affiliation(s)
- Hong-Jing Zhu
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun, China
| | - Ying-Ying Sun
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun, China
| | - Yijing Du
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130021, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P.R. China
| | - Sheng-Yu Zhou
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun, China
| | - Yang Qu
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun, China
| | - Shu-Yan Pang
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun, China
| | - Shoujun Zhu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130021, P.R. China.
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P.R. China.
| | - Yi Yang
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun, China.
| | - Zhen-Ni Guo
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun, China.
- Neuroscience Research Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun, China.
| |
Collapse
|
6
|
Vinícius-Araújo M, Shrivastava N, Silva Loures G, Krause RF, Sousa MH, de Santana RC, Bakuzis AF. Integration of 3D Fluorescence Imaging and Luminescent Thermometry with Core-Shell Engineered NaYF 4:Nd 3+/Yb 3+/Ho 3+ Nanoparticles. Inorg Chem 2024; 63:1840-1852. [PMID: 38232297 DOI: 10.1021/acs.inorgchem.3c03410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
The design of rare-earth-doped upconversion/downshifting nanoparticles (NPs) for theoretical use in nanomedicine has garnered considerable interest. Previous research has emphasized luminescent nanothermometry and photothermal therapy, while three-dimensional (3D) near-infrared (NIR) luminescent tracers have received less attention. Our study introduces Nd3+-, Yb3+-, and Ho3+-doped NaYF4 core-shell luminescent NPs as potential multiparametric nanothermometers and NIR imaging tracers. Nd3+ sensitizes at 804 nm, while Yb3+ bridges to activators Ho3+. We evaluated the photoluminescence properties of Nd3+-, Yb3+-, and Ho3+-doped core and core-shell NPs synthesized via polyol-mediated and thermal decomposition methods. The NaYF4:NdYbHo(7/15/3%)@NaYF4:Nd(15%) core-shell NPs demonstrate competitive nanothermometry capabilities. Specifically, the polyol-synthesized sample exhibits a sensitivity of 0.27% K-1 at 313 K (40 °C), whereas the thermally decomposed synthesized sample shows a significantly higher sensitivity of 0.55% K-1 at 313 K (40 °C) in the near-infrared range. Control samples indicate back energy transfer processes from both Yb and Ho to Nd, while Yb to Ho energy transfer enhances Ho3+-driven upconversion transitions in green and red wavelengths, suggesting promise for photodynamic therapy. Fluorescence molecular tomography confirms 3D NIR fluorescence nanoparticle localization in a biological media after injection, highlighting the potential of core-shell NPs as NIR luminescent tracers. The strategy's clinical impact lies in photothermal treatment planning, leveraging core-shell NPs for (pre)clinical applications, and enabling the easy addition of new functionalities through distinct ion doping.
Collapse
Affiliation(s)
| | - Navadeep Shrivastava
- Department of Chemistry, Physics and Materials Science, Fayetteville State University, Fayetteville, North Carolina 28301, United States
| | | | - Rafael Freire Krause
- Institute of Physics, Federal University of Goiás, Goiânia, GO 74690-900, Brazil
| | | | | | - Andris Figueiroa Bakuzis
- Institute of Physics, Federal University of Goiás, Goiânia, GO 74690-900, Brazil
- CNanoMed, Federal University of Goiás, Goiânia, GO 74690-631, Brazil
| |
Collapse
|
7
|
Liu YL, Wang TH, Yeh NT, Huang WJ, Tzang BS, Wu IT, Chin HY, Hu SH, Hsu TC, Chiang WH. Tumor-activated targetable photothermal chemotherapy using IR780/zoledronic acid-containing hybrid polymeric nanoassemblies with folate modification to treat aggressive breast cancer. NANOSCALE 2024; 16:1415-1427. [PMID: 38167914 DOI: 10.1039/d3nr05637f] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
To effectively treat aggressive breast cancer by tumor-activated targetable photothermal chemotherapy, in this work, folate (FA)-modified hybrid polymeric nanoassemblies (HPNs) with a poly(ethylene glycol) (PEG)-detachable capability are developed as vehicles for tumor-targeted co-delivery of IR780, a lipophilic photothermal reagent, and zoledronic acid (ZA), a hydrophilic chemotherapy drug. Through hydrophobic interaction-induced co-assembly, IR780 molecules and ZA/poly(ethylenimine) (PEI) complexes were co-encapsulated into a poly(lactic-co-glycolic acid) (PLGA)-rich core stabilized by the amphiphilic FA-modified D-α-tocopheryl poly(ethylene glycol) succinate (FA-TPGS) and acidity-sensitive PEG-benzoic imine-octadecane (C18) (PEG-b-C18) conjugates. The developed FA-ZA/IR780@HPNs with high ZA and IR780 payloads not only showed excellent colloidal stability in a serum-containing milieu, but also promoted IR780-based photostability and photothermal conversion efficiency. Furthermore, for FA-ZA/IR780@HPNs under simulated physiological conditions, the premature leakage of IR780 and ZA molecules was remarkably declined. In a mimetic acidic tumor microenvironment, the uptake of FA-ZA/IR780@HPNs by FA receptor-overexpressed 4T1 breast cancer cells was remarkably promoted by PEG detachment combined with FA receptor-mediated endocytosis, thus effectively hindering migration of cancer cells and augmenting the anticancer efficacy of photothermal chemotherapy. Notably, the in vivo studies demonstrated that the FA-ZA/IR780@HPNs largely deposited at 4T1 tumor sites and profoundly suppressed tumor growth and metastasis without severe systemic toxicity upon near infrared (NIR)-triggered IR780-mediated hyperthermia integrated with ZA chemotherapy. This work presents a practical strategy to treat aggressive breast tumors with tumor-triggered targetable photothermal chemotherapy using FA-ZA/IR780@HPNs.
Collapse
Affiliation(s)
- Yu-Ling Liu
- Department of Chemical Engineering, National Chung Hsing University, Taichung 402, Taiwan.
| | - Tzu-Hao Wang
- Department of Chemical Engineering, National Chung Hsing University, Taichung 402, Taiwan.
| | - Nien-Tzu Yeh
- Department of Chemical Engineering, National Chung Hsing University, Taichung 402, Taiwan.
| | - Wei-Jen Huang
- Department of Chemical Engineering, National Chung Hsing University, Taichung 402, Taiwan.
| | - Bor-Show Tzang
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan.
- Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
- Immunology Research Center, Chung Shan Medical University, Taichung 402, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - I-Ting Wu
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan.
| | - Hao-Yang Chin
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan.
| | - Shang-Hsiu Hu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Tsai-Ching Hsu
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan.
- Immunology Research Center, Chung Shan Medical University, Taichung 402, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Wen-Hsuan Chiang
- Department of Chemical Engineering, National Chung Hsing University, Taichung 402, Taiwan.
| |
Collapse
|
8
|
Immunogenic Cell Death Photothermally Mediated by Erythrocyte Membrane-Coated Magnetofluorescent Nanocarriers Improves Survival in Sarcoma Model. Pharmaceutics 2023; 15:pharmaceutics15030943. [PMID: 36986804 PMCID: PMC10051374 DOI: 10.3390/pharmaceutics15030943] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/03/2023] [Accepted: 03/12/2023] [Indexed: 03/17/2023] Open
Abstract
Inducing immunogenic cell death (ICD) during cancer therapy is a major challenge that might significantly improve patient survival. The purpose of this study was to develop a theranostic nanocarrier, capable both of conveying a cytotoxic thermal dose when mediating photothermal therapy (PTT) after its intravenous delivery, and of consequently inducing ICD, improving survival. The nanocarrier consists of red blood cell membranes (RBCm) embedding the near-infrared dye IR-780 (IR) and camouflaging Mn-ferrite nanoparticles (RBCm-IR-Mn). The RBCm-IR-Mn nanocarriers were characterized by size, morphology, surface charge, magnetic, photophysical, and photothermal properties. Their photothermal conversion efficiency was found to be size- and concentration-dependent. Late apoptosis was observed as the cell death mechanism for PTT. Calreticulin and HMGB1 protein levels increased for in vitro PTT with temperature around 55 °C (ablative regime) but not for 44 °C (hyperthermia), suggesting ICD elicitation under ablation. RBCm-IR-Mn were then intravenously administered in sarcoma S180-bearing Swiss mice, and in vivo ablative PTT was performed five days later. Tumor volumes were monitored for the subsequent 120 days. RBCm-IR-Mn-mediated PTT promoted tumor regression in 11/12 animals, with an overall survival rate of 85% (11/13). Our results demonstrate that the RBCm-IR-Mn nanocarriers are great candidates for PTT-induced cancer immunotherapy.
Collapse
|
9
|
Muhammad Usama S, Gao Z, Arancillo M, Burgess K. Cytotoxicities of Tumor-Seeking Dyes: Impact on Future Clinical Trials. ChemMedChem 2023; 18:e202200561. [PMID: 36630600 PMCID: PMC10010615 DOI: 10.1002/cmdc.202200561] [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: 10/17/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023]
Abstract
Heptamethine (Cy7) dyes with meso-Cl substituents injected intravenously (iv) into mice accumulate in tumors and persist there over several days. We believe this occurs via meso-Cl displacement by the only free cysteine residues of albumin; therefore, conjugating tumor-seeking dyes with fragments can increase selective therapeutic delivery to tumors and drug residence. This strategy has elevated significance recently because the first tumor-seeking dye-drug conjugate has moved into clinical trials. Options for further clinical research include modifying the dye, and use of preformed albumin adducts instead of dyes alone. Herein we show correlations of cytotoxicities, lipophilicities, organelle localization, apoptosis, cell-cycle arrest, wound healing/migration assays, and reactivities/affinities with human serum albumin are difficult to observe. However, our studies arrived at an important conclusion: preformed dye-drug-HSA adducts are less cytotoxic, and therefore preferable for subsequent clinical work, relative to direct injection of meso-Cl-containing forms.
Collapse
Affiliation(s)
- Syed Muhammad Usama
- Department of Chemistry, Texas A & M University, Box 30012, College Station, TX 77842, USA
| | - Zhe Gao
- Department of Chemistry, Texas A & M University, Box 30012, College Station, TX 77842, USA
| | - Maritess Arancillo
- Department of Chemistry, Texas A & M University, Box 30012, College Station, TX 77842, USA
| | - Kevin Burgess
- Department of Chemistry, Texas A & M University, Box 30012, College Station, TX 77842, USA
| |
Collapse
|
10
|
Barcelos JM, Hayasaki TG, de Santana RC, Lima EM, Mendanha SA, Bakuzis AF. Photothermal Properties of IR-780-Based Nanoparticles Depend on Nanocarrier Design: A Comparative Study on Synthetic Liposomes and Cell Membrane and Hybrid Biomimetic Vesicles. Pharmaceutics 2023; 15:pharmaceutics15020444. [PMID: 36839765 PMCID: PMC9961772 DOI: 10.3390/pharmaceutics15020444] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/19/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Biomimetic nanoparticles hold great promise for photonic-mediated nanomedicine due to the association of the biological functionality of the membrane with the physical/chemical goals of organic/inorganic structures, but studies involving fluorescent biomimetic vesicles are still scarce. The purpose of this article is to determine how photothermal therapy (PTT) with theranostic IR-780-based nanoparticles depends on the dye content, cholesterol content, lipid bilayer phase and cell membrane type. The photophysical responses of synthetic liposomes, cell membrane vesicles and hybrid nanoparticles are compared. The samples were characterized by nanoparticle tracking analysis, photoluminescence, electron spin resonance, and photothermal- and heat-mediated drug release experiments, among other techniques. The photothermal conversion efficiency (PCE) was determined using Roper's method. All samples excited at 804 nm showed three fluorescence bands, two of them independent of the IR-780 content. Samples with a fluorescence band at around 850 nm showed photobleaching (PBL). Quenching was higher in cell membrane vesicles, while cholesterol inhibited quenching in synthetic liposomes with low dye content. PTT depended on the cell membrane and was more efficient for melanoma than erythrocyte vesicles. Synthetic liposomes containing cholesterol and a high amount of IR-780 presented superior performance in PTT experiments, with a 2.4-fold PCE increase in comparison with free IR-780, no PBL and the ability to heat-trigger doxorubicin release.
Collapse
Affiliation(s)
- Júlia Muniz Barcelos
- Institute of Physics, Federal University of Goiás, Goiânia 74690-900, GO, Brazil
| | | | | | - Eliana Martins Lima
- Farmatec, School of Pharmacy, Federal University of Goiás, Goiânia 74690-631, GO, Brazil
- CNanomed, Federal University of Goiás, Goiânia 74690-631, GO, Brazil
| | - Sebastião Antonio Mendanha
- Institute of Physics, Federal University of Goiás, Goiânia 74690-900, GO, Brazil
- Farmatec, School of Pharmacy, Federal University of Goiás, Goiânia 74690-631, GO, Brazil
- CNanomed, Federal University of Goiás, Goiânia 74690-631, GO, Brazil
| | - Andris Figueiroa Bakuzis
- Institute of Physics, Federal University of Goiás, Goiânia 74690-900, GO, Brazil
- CNanomed, Federal University of Goiás, Goiânia 74690-631, GO, Brazil
- Correspondence:
| |
Collapse
|
11
|
Li M, Zhang Y, Ma J, Du J. Albumin-based nanoparticle for dual-modality imaging of the lymphatic system. RSC Adv 2023; 13:2248-2255. [PMID: 36741156 PMCID: PMC9838117 DOI: 10.1039/d2ra07414a] [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: 11/22/2022] [Accepted: 12/27/2022] [Indexed: 01/15/2023] Open
Abstract
The lymphatic system is a complex network of lymphatic vessels, lymph nodes, and lymphoid organs. The current understanding of the basic mechanism and framework of the lymphatic system is relatively limited and not ideal for exploring the function of the lymphatic system, diagnosing lymphatic system diseases, and controlling tumor metastasis. Imaging modalities for evaluating lymphatic system diseases mainly include lymphatic angiography, reactive dye lymphatic angiography, radionuclide lymphatic angiography, computed tomography, and ultrasonography. However, these are insufficient for clinical diagnosis. Some novel imaging methods, such as magnetic resonance imaging, positron emission computed tomography, single-photon emission computed tomography, contrast-enhanced ultrasonography, and near-infrared imaging with agents such as cyanine dyes, can reveal lymphatic system information more accurately and in detail. We fabricated an albumin-based fluorescent probe for dual-modality imaging of the lymphatic system. A near-infrared cyanine dye, IR-780, was absorbed into bovine serum albumin (BSA), which was covalently linked to a molecule of diethylenetriaminepentaacetic acid to chelate gadolinium Gd3+. The fabricated IR-780@BSA@Gd3+ nanocomposite demonstrates strong fluorescence and high near-infrared absorption and can be used as a T1 contrast agent for magnetic resonance imaging. In vivo dual-modality fluorescence and magnetic resonance imaging showed that IR-780@BSA@Gd3+ rapidly returned to the heart through the lymphatic circulation after it was injected into the toe webs of mice, facilitating good lymphatic imaging. The successful fabrication of the new IR-780@BSA@Gd3+ nanocomposite will facilitate the study of the mechanism and morphological structure of the lymphatic system.
Collapse
Affiliation(s)
- Mingze Li
- Jilin Provincial Key Laboratory of Lymphatic Surgical Disease, Engineering Laboratory of Lymphatic Surgery Jilin Province, China-Japan Union Hospital of Jilin UniversityChangchunJilin130031P. R. China
| | - Yundong Zhang
- Jilin Provincial Key Laboratory of Lymphatic Surgical Disease, Engineering Laboratory of Lymphatic Surgery Jilin Province, China-Japan Union Hospital of Jilin UniversityChangchunJilin130031P. R. China
| | - Jinli Ma
- Jilin Provincial Key Laboratory of Lymphatic Surgical Disease, Engineering Laboratory of Lymphatic Surgery Jilin Province, China-Japan Union Hospital of Jilin UniversityChangchunJilin130031P. R. China
| | - Jianshi Du
- Jilin Provincial Key Laboratory of Lymphatic Surgical Disease, Engineering Laboratory of Lymphatic Surgery Jilin Province, China-Japan Union Hospital of Jilin UniversityChangchunJilin130031P. R. China
| |
Collapse
|
12
|
Xia Y, Wu Y, Cao J, Wang J, Chen Z, Li C, Zhang X. Liposomal Glucose Oxidase for Enhanced Photothermal Therapy and Photodynamic Therapy against Breast Tumors. ACS Biomater Sci Eng 2022; 8:1892-1906. [PMID: 35404565 DOI: 10.1021/acsbiomaterials.1c01311] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Organic near-infrared fluorescent dye mediated photothermal therapy (PTT) and photodynamic therapy (PDT) suffer from heat shock response, since, heat shock proteins (HSPs) are overexpressed and can repair the proteins damaged by PTT and PDT. Starvation therapy by glucose oxide (GOx) can inhibit the heat shock response by limiting the energy supply. However, the delivery of sufficient and active GOx remains a challenge. To solve this problem, we utilize liposomes as drug carriers and prepare GOx loaded liposome (GOx@Lipo) with a high drug loading content (12.0%) and high enzymatic activity. The successful delivery of GOx shows excellent inhibition of HSPs and enhances PTT and PDT. Additionally, we apply the same liposome formulation to load near-infrared dye 1,1'-dioctadecyl-3,3,3',3'-tetramethylindotricarbo cyanine iodide (DiR) and prepare DiR contained liposomes (DiR@Lipo) for PTT and PDT. The liposomal formulation substantially enhances the PTT and PDT properties of DiR as well as the cellular uptake and tumor accumulation. Finally, the combination therapy shows excellent tumor inhibition on 4T1 tumor-bearing mice. Interestingly, we also find that the starvation therapy can efficiently inhibit tumor metastasis, which is probably due to the immunogenic effect. Our work presents a biocompatible and effective carrier for the combination of starvation therapy and phototherapy, emphasizing the importance of auxiliary starvation therapy against tumor metastasis and offering important guidance for clinical PTT and PDT.
Collapse
Affiliation(s)
- Yuqiong Xia
- Engineering Research Center of Molecular- and Neuro-Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710026, China
| | - Yankun Wu
- Engineering Research Center of Molecular- and Neuro-Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710026, China
| | - Jianxia Cao
- Engineering Research Center of Molecular- and Neuro-Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710026, China
| | - Jun Wang
- Engineering Research Center of Molecular- and Neuro-Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710026, China
| | - Zhaoxu Chen
- Engineering Research Center of Molecular- and Neuro-Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710026, China
| | - Cairu Li
- Engineering Research Center of Molecular- and Neuro-Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710026, China
| | - Xianghan Zhang
- Engineering Research Center of Molecular- and Neuro-Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710026, China
| |
Collapse
|
13
|
Homologous targeting nanoparticles for enhanced PDT against osteosarcoma HOS cells and the related molecular mechanisms. J Nanobiotechnology 2022; 20:83. [PMID: 35177075 PMCID: PMC8851855 DOI: 10.1186/s12951-021-01201-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 12/09/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND No prominent advancements in osteosarcoma (OS) treatment have been made in the past 20 years. Although photodynamic therapy (PDT) is an emerging technique for cancer therapy, the lack of targeted photosensitizers for OS treatment severely limits its applications. RESULTS In this study, we constructed a potential theranostic nanoplatform by using (poly (lactic-co-glycolic) acid (PLGA) nanoparticles (NPs) encapsulating IR780 into the shell (PLGA-IR780 NPs), which were further camouflaged with human OS cell membranes from the HOS cell line (MH-PLGA-IR780 NPs). These constructed NPs showed the capacity for homologous targeting with excellent photoacoustic (PA)/fluorescence (FL) imaging ability. Benefitting from their homologous targeting capacity, MH-PLGA-IR780 NPs obviously promoted cell endocytosis in vitro and tumor accumulation in vivo, which could further improve PDT performance under near-infrared (NIR) irradiation. In addition, to their homologous targeting and PA/FL dual-mode imaging ability, MH-PLGA-IR780 NPs had advantages in penetrating deeper into tumor tissues and in real-time dynamic distribution monitoring in vivo, which laid a foundation for further clinical applications in OS. Moreover, we demonstrated that PDT guided by the constructed NPs could significantly induce HOS cells apoptosis and ferroptosis via excessive accumulation of reactive oxygen species (ROS), and further determined that the potential anticancer molecular mechanism of apoptosis was triggered by the release of cytochrome c-activated mitochondrial apoptosis (endogenous apoptosis), and that ferroptosis caused the activation of nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy and the inactivation of glutathione peroxidase 4 (GPX4), synergistically leading to excessive accumulation of Lipid-ROS and Lipid peroxides (LPOs). Concurrently, MH-PLGA-IR780 NPs-guided PDT also showed an obvious inhibitory effect on tumor growth in vivo. CONCLUSION These results suggest that this homologous targeting-based theranostic nanoplatform provides an effective method to improve PDT performance in OS and contributes a new and promising approach for OS therapy.
Collapse
|
14
|
Prospero AG, Buranello LP, Fernandes CA, Dos Santos LD, Soares G, C Rossini B, Zufelato N, Bakuzis AF, de Mattos Fontes MR, de Arruda Miranda JR. Corona protein impacts on alternating current biosusceptometry signal and circulation times of differently coated MnFe 2O 4 nanoparticles. Nanomedicine (Lond) 2021; 16:2189-2206. [PMID: 34533056 DOI: 10.2217/nnm-2021-0195] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: We evaluated the impacts of corona protein (CP) formation on the alternating current biosusceptometry (ACB) signal intensity and in vivo circulation times of three differently coated magnetic nanoparticles (MNP): bare, citrate-coated and bovine serum albumin-coated MNPs. Methods: We employed the ACB system, gel electrophoresis and mass spectrometry analysis. Results: Higher CP formation led to a greater reduction in the in vitro ACB signal intensity and circulation time. We found fewer proteins forming the CP for the bovine serum albumin-coated MNPs, which presented the highest circulation time in vivo among the MNPs studied. Conclusion: These data showed better biocompatibility, stability and magnetic signal uniformity in biological media for bovine serum albumin-coated MNPs than for citrate-coated MNPs and bare MNPs.
Collapse
Affiliation(s)
- Andre Gonçalves Prospero
- Department of Biophysics and Pharmacology, São Paulo State University, Botucatu, São Paulo, 18618-689, Brazil
| | - Lais Pereira Buranello
- Department of Biophysics and Pharmacology, São Paulo State University, Botucatu, São Paulo, 18618-689, Brazil
| | - Carlos Ah Fernandes
- Department of Biophysics and Pharmacology, São Paulo State University, Botucatu, São Paulo, 18618-689, Brazil.,Museum National d'Histoire Naturelle, Institut de Minéralogie, Physique des Matériaux et Cosmochimie, IMPMC, Sorbonne Université, UMR 7590, CNRS, Paris, France
| | - Lucilene Delazari Dos Santos
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo State University (UNESP), Botucatu, São Paulo, 18618-687, Brazil.,Biotechnology Institute, São Paulo State University, Botucatu, São Paulo, 18607-440, Brazil
| | - Guilherme Soares
- Department of Biophysics and Pharmacology, São Paulo State University, Botucatu, São Paulo, 18618-689, Brazil
| | - Bruno C Rossini
- Biotechnology Institute, São Paulo State University, Botucatu, São Paulo, 18607-440, Brazil
| | - Nícholas Zufelato
- Institute of Physics and CNanoMed, Federal University of Goiás, Goiânia, 74690-900, Brazil
| | | | - Marcos R de Mattos Fontes
- Department of Biophysics and Pharmacology, São Paulo State University, Botucatu, São Paulo, 18618-689, Brazil
| | - José R de Arruda Miranda
- Department of Biophysics and Pharmacology, São Paulo State University, Botucatu, São Paulo, 18618-689, Brazil
| |
Collapse
|
15
|
Abstract
Active targeting uses molecular fragments that bind receptors overexpressed on cell surfaces to deliver cargoes, and this selective delivery to diseased over healthy tissue is valuable in diagnostic imaging and therapy. For instance, targeted near-infrared (near-IR) dyes can mark tissue to be excised in surgery, and radiologists can use active targeting to concentrate agents for positron emission tomography (PET) in tumor tissue to monitor tumor metastases. Selective delivery to diseased tissue is also valuable in some treatments wherein therapeutic indexes (toxic/effective doses) are key determinants of efficacy. However, active targeting will only work for cells expressing the pivotal cell surface receptor that is targeted. That is a problem because tumors, even ones derived from the same organ, are not homogeneous, patient-to-patient variability is common, and heterogeneity can occur even in the same patient, so monotherapy with one actively targeted agent is unlikely to be uniformly effective. A particular category of fluorescent heptamethine cyanine-7 (Cy-7) dyes, here called tumor seeking dyes, offer a way to circumvent this problem because they selectively accumulate in any solid tumor. Furthermore, they persist in tumor tissue for several days, sometimes longer than 72 h. Consequently, tumor seeking dyes are near-IR fluorescent targeting agents that, unlike mAbs (monoclonal antibodies), accumulate in any solid lesion, thus overcoming tumor heterogeneity, and persist there for long periods, circumventing the rapid clearance problems that bedevil low molecular mass drugs. Small molecule imaging agents and drugs attached to tumor-seeking dyes have high therapeutic indices and long residence times in cancer cells and tumor tissue. All this sounds too good to be true. We believe most of this is true, but the controversy is associated with how and why these characteristics arise. Prior to our studies, the prevailing hypothesis, often repeated, was that tumor seeking dyes are uptaken by organic anion transporting polypeptides (OATPs) overexpressed on cancer cells. This Account summarizes evidence indicating tumor seeking Cy-7 dyes have exceptional accumulation and persistence properties because they covalently bind to albumin in vivo. That adduct formation provides a convenient way to form albumin-bound pharmaceuticals labeled with near-IR fluorophores which can be tracked in vivo. This understanding may facilitate more rapid developments of generally applicable actively targeted reagents.
Collapse
Affiliation(s)
- Syed Muhammad Usama
- Department of Chemistry, Texas A&M University, Box 30012, College Station, Texas 77842, United States
| | - Kevin Burgess
- Department of Chemistry, Texas A&M University, Box 30012, College Station, Texas 77842, United States
| |
Collapse
|
16
|
Capistrano G, Rodrigues HF, Zufelato N, Gonçalves C, Cardoso CG, Silveira-Lacerda EP, Bakuzis AF. Noninvasive intratumoral thermal dose determination during in vivo magnetic nanoparticle hyperthermia: combining surface temperature measurements and computer simulations. Int J Hyperthermia 2021; 37:120-140. [PMID: 33426991 DOI: 10.1080/02656736.2020.1826583] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
PURPOSE Noninvasive thermometry during magnetic nanoparticle hyperthermia (MNH) remains a challenge. Our pilot study proposes a methodology to determine the noninvasive intratumoral thermal dose during MNH in the subcutaneous tumor model. METHODS Two groups of Ehrlich bearing-mice with solid and subcutaneous carcinoma, a control group (n = 6), and a MNH treated group (n = 4) were investigated. Histopathology was used to evaluate the percentage of non-viable lesions in the tumor. MNH was performed at 301 kHz and 17.5 kA.m-1, using a multifunctional nanocarrier. Surface temperature measurements were obtained using an infrared camera, where an ROI with 750 pixels was used for comparison with computer simulations. Realistic simulations of the bioheat equation were obtained by combining histopathology intratumoral lesion information and surface temperature agreement of at least 50% of the pixel's temperature data calculated and measured at the surface. RESULTS One animal of the MNH group showed tumor recurrence, while two others showed complete tumor remission (monitored for 585 days). Sensitivity analysis of the simulation parameters indicated low tumor blood perfusion. Numerical simulations indicated, for the animals with complete remission, an irreversible tissue injury of 91 ± 5% and 100%, while the one with recurrence had a lower value, 56 ± 7%. The computer simulations also revealed the in vivo heat efficiency of the nanocarrier. CONCLUSION A new methodology for determining noninvasively the three-dimensional intratumoral thermal dose during MNH was developed. The method demonstrates the potential for predicting the long-term preclinical outcome of animals treated with MNH.
Collapse
Affiliation(s)
- Gustavo Capistrano
- Instituto de Física, Universidade Federal de Goiás, Goiânia, Brazil.,Instituto Federal de Mato Grosso, Pontes e Lacerda, Brazil
| | - Harley F Rodrigues
- Instituto de Física, Universidade Federal de Goiás, Goiânia, Brazil.,Instituto Federal de Goiás, Curso de Licenciatura em Física, Goiânia, Brazil
| | | | - Cristhiane Gonçalves
- Instituto de Física, Universidade Federal de Goiás, Goiânia, Brazil.,Universidade Tecnológica Federal do Paraná, Ponta Grossa, Brazil
| | - Clever G Cardoso
- Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | | | - Andris F Bakuzis
- Instituto de Física, Universidade Federal de Goiás, Goiânia, Brazil
| |
Collapse
|
17
|
Keerthiga R, Zhao Z, Pei D, Fu A. Photodynamic Nanophotosensitizers: Promising Materials for Tumor Theranostics. ACS Biomater Sci Eng 2020; 6:5474-5485. [PMID: 33320544 DOI: 10.1021/acsbiomaterials.0c01058] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Photodynamic theranostics/therapy (PDT) is a potential strategy for selectively imaging malignant sites and treating cancer via a non-invasive therapeutic method. Photosensitizers, the crucial components of PDT, enable colocalization of photons and light, and photon/light therapy in the therapeutic window of 400-900 nm exhibits photocytotoxicity to tumor cells. Due to their high biostability and photocytotoxicity, nanophotosensitizers (NPSs) are of much interest for malignant tumor theranostics at present. NPS-activated photons transfer energy through the absorption of a photon and convert molecular oxygen to the singlet reactive oxygen species, which leads to apoptosis and necrosis. Moreover, NPSs modified by polymers, including PLGA, PEG-PLA, PDLLA, PVCL-g-PLA, and P(VCL-co-VIM)-g-PLA, exhibit excellent biocompatibility, and a tumor-targeting molecule linked on the nanoparticle surface can precisely deliver NPSs into the tumor region. The development of NPSs will accelerate the progress in tumor theranostics through the photon/light pathway.
Collapse
Affiliation(s)
- Rajendiran Keerthiga
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zizhen Zhao
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Desheng Pei
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Ailing Fu
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|