1
|
Li Z, Liu J, Hu Y, Li Z, Fan X, Sun Y, Besenbacher F, Chen C, Yu M. Biocompatible PEGylated bismuth nanocrystals: "All-in-one" theranostic agent with triple-modal imaging and efficient in vivo photothermal ablation of tumors. Biomaterials 2017; 141:284-295. [PMID: 28709019 DOI: 10.1016/j.biomaterials.2017.06.033] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 05/26/2017] [Accepted: 06/22/2017] [Indexed: 11/28/2022]
Abstract
Biocompatible single-component theranostic agents integrating multimodal imaging and therapeutic functions (namely, "all-in one" agents) are highly desired for clinical cancer treatments. Herein, PEGylated pure metallic bismuth nanocrystals (Bi-PEG NCs) have been developed to be a competent theranostic agent for in vivo high-performance multimodal bio-imaging and photothermal ablation of tumors. The resultant Bi-PEG NCs show excellent physiological stability, biocompatibility, prolonged blood circulation half-life and preferential tumor accumulation. Thanking to the strong near-infrared (NIR) absorbance as well as the high photothermal conversion efficiency and conversion stability, highly effective in vivo photothermal ablation on tumors has been realized upon NIR irradiation, without noticeable toxicity. Impressively, the Bi-PEG NCs show ultrahigh X-ray computed topography (CT) enhancement efficiency (∼60.3 HU mL mg-1), overwhelming all CT contrast agents reported so far. Combining the strong CT contrast ability and photoacoustic/photothermal effect, high-contrast CT, photoacoustic (PA) and infrared thermal (IRT) triple-modal imaging have been demonstrated both in vitro and in vivo. This work highlights the potentials of such NCs as a powerful "all-in-one" theranostic nanoplatform for bioimaging and antitumor therapy, and may have provided a rather promising candidate for clinically-applied antitumor treatments based on single-component agents.
Collapse
|
Journal Article |
8 |
59 |
2
|
Zhao Z, Xu K, Fu C, Liu H, Lei M, Bao J, Fu A, Yu Y, Zhang W. Interfacial engineered gadolinium oxide nanoparticles for magnetic resonance imaging guided microenvironment-mediated synergetic chemodynamic/photothermal therapy. Biomaterials 2019; 219:119379. [PMID: 31376746 DOI: 10.1016/j.biomaterials.2019.119379] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 07/16/2019] [Accepted: 07/24/2019] [Indexed: 12/12/2022]
Abstract
Engineering interfacial structure of biomaterials have drawn much attention due to it can improve the diagnostic accuracy and therapy efficacy of nanomedicine, even introducing new moiety to construct theranostic agents. Nanosized magnetic resonance imaging contrast agent holds great promise for the clinical diagnosis of disease, especially tumor and brain disease. Thus, engineering its interfacial structure can form new theranostic platform to achieve effective disease diagnosis and therapy. In this study, we engineered the interfacial structure of typical MRI contrast agent, Gd2O3, to form a new theranostic agent with improved relaxivity for MRI guided synergetic chemodynamic/photothermal therapy. The synthesized Mn doped gadolinium oxide nanoplate exhibit improved T1 contrast ability due to large amount of efficient paramagnetic metal ions and synergistic enhancement caused by the exposed Mn and Gd cluster. Besides, the introduced Mn element endow this nanomedicine with the Fenton-like ability to generate OH from excess H2O2 in tumor site to achieve chemodynamic therapy (CDT). Furthermore, polydopamine engineered surface allow this nanomedicine with effective photothermal conversion ability to rise local temperature and accelerate the intratumoral Fenton process to achieve synergetic CDT/photothermal therapy (PTT). This work provides new guidance for designing magnetic resonance imaging guided synergetic CDT/PTT to achieve tumor detection and therapy.
Collapse
|
Research Support, Non-U.S. Gov't |
6 |
51 |
3
|
Jing L, Liang X, Li X, Yang Y, Dai Z. Covalent attachment of Mn-porphyrin onto doxorubicin-loaded poly(lactic acid) nanoparticles for potential magnetic resonance imaging and pH-sensitive drug delivery. Acta Biomater 2013; 9:9434-41. [PMID: 23962645 DOI: 10.1016/j.actbio.2013.08.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 07/31/2013] [Accepted: 08/12/2013] [Indexed: 01/30/2023]
Abstract
In this paper, theranostic nanoparticles (MnP-DOX NPs) were fabricated by conjugating Mn-porphyrin onto the surface of doxorubicin (DOX)-loaded poly(lactic acid) (PLA) nanoparticles (DOX NPs) for potential T1 magnetic resonance imaging and pH-sensitive drug delivery. An in vitro drug release study showed that the release rate of DOX from MnP-DOX NPs was slow at neutral pH but accelerated significantly in acidic conditions. It was found that MnP-DOX NPs could be easily internalized by HeLa cells and effectively suppressed the growth of HeLa cells and HT-29 cells due to the accelerated drug release in acidic lysosomal compartments. Magnetic resonance imaging (MRI) scanning analysis demonstrated that MnP-DOX NPs had much higher longitudinal relaxivity in water (r1 value of 27.8 mM(-1) s(-1) of Mn(3+)) than Mn-porphyrin (Mn(III)TPPS3NH2; r1 value of 6.70 mM(-1) s(-1) of Mn(3+)), behaving as an excellent contrast agent for T1-weighted MRI both in vitro and in vivo. In summary, such a smart and promising nanoplatform integrates multiple capabilities for effective cancer diagnosis and therapy.
Collapse
|
Research Support, Non-U.S. Gov't |
12 |
46 |
4
|
Progresses in polymeric nanoparticles for delivery of tyrosine kinase inhibitors. Life Sci 2021; 278:119642. [PMID: 34033837 DOI: 10.1016/j.lfs.2021.119642] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 05/07/2021] [Accepted: 05/18/2021] [Indexed: 02/07/2023]
Abstract
Tyrosine kinase inhibitors (TKIs), as an important class of chemotherapeutic drugs, induce apoptosis by altering the path of the cellular signal, resulting in cell death. However, some chemotherapeutic drugs have a limited therapeutic index and are usually destructive as well as unpredictable. In addition, the limitation of early diagnosis and inefficiency of some of the drugs in ordinary treatments lead to disease progression and decreases in the survival of cancer patients. For this purpose, various methods have been proposed, among them, nanomedicine has transpired as a modern approach for the treatment of multiple cancers. Over the last two decades, targeted therapy has been developed for cancer-specific cells/tissues and has rather restricted nonselective toxicities. In vivo and in vitro studies demonstrated nanoparticles (NPs), nano-scale drugs, and nano-carriers alone or in combination with other therapeutic, imaging, and theranostic agents would be applied as an effective approach targeting a diversity of malignant tissue. Therefore, using the latest advances in materials science and biomaterials, biology, it has happened that general diagnosis and treatment can be performed. In this review, we indicated the applications of theranostic nano-polymer and nano-liposome to TKIs delivery.
Collapse
|
Review |
4 |
38 |
5
|
Dong Q, Yang H, Wan C, Zheng D, Zhou Z, Xie S, Xu L, Du J, Li F. Her2-Functionalized Gold-Nanoshelled Magnetic Hybrid Nanoparticles: a Theranostic Agent for Dual-Modal Imaging and Photothermal Therapy of Breast Cancer. NANOSCALE RESEARCH LETTERS 2019; 14:235. [PMID: 31448377 PMCID: PMC6709082 DOI: 10.1186/s11671-019-3053-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/17/2019] [Indexed: 05/08/2023]
Abstract
Targeted theranostic platform that integrates multi-modal imaging and therapeutic function is emerging as a promising strategy for earlier detection and precise treatment of cancer. Herein, we designed targeted gold-nanoshelled poly (lactic-co-glycolic acid) (PLGA) magnetic hybrid nanoparticles carrying anti-human epidermal growth factor receptor 2 (Her2) antibodies (Her2-GPH NPs) for dual-modal ultrasound (US)/magnetic resonance (MR) imaging and photothermal therapy of breast cancer. The agent was fabricated by coating gold nanoshell around PLGA nanoparticles co-loaded with perfluorooctyl bromide (PFOB) and superparamagnetic iron oxide nanoparticles (SPIOs), followed by conjugating with anti-Her2 antibodies. Cell-targeting studies demonstrated receptor-mediated specific binding of the agent to Her2-positive human breast cancer SKBR3 cells, and its binding rate was significantly higher than that of Her2-negative cells (P < 0.001). In vitro, the agent had capabilities for contrast-enhanced US imaging as well as T2-weighted MR imaging with a relatively high relaxivity (r2 = 441.47 mM-1 s-1). Furthermore, the Her2 functionalization of the agent prominently enhanced the US/MR molecular imaging effect of targeted cells by cell-specific binding. Live/dead cell assay and targeted photothermal cytotoxicity experiments confirmed that Her2-GPH NPs could serve as effective photoabsorbers to specifically induce SKBR3 cell death upon near-infrared laser irradiation. In summary, Her2-GPH NPs were demonstrated to be novel targeted theranostic agents with great potential to facilitate early non-invasive diagnosis and adjuvant therapy of breast cancer.
Collapse
|
brief-report |
6 |
35 |
6
|
Li Q, Chen K, Huang W, Ma H, Zhao X, Zhang J, Zhang Y, Fang C, Nie L. Minimally invasive photothermal ablation assisted by laparoscopy as an effective preoperative neoadjuvant treatment for orthotopic hepatocellular carcinoma. Cancer Lett 2020; 496:169-178. [PMID: 32987139 DOI: 10.1016/j.canlet.2020.09.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 08/23/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023]
Abstract
Nanoparticle-based photothermal ablation (PTA) has been intensively investigated recently. However, the poor biocompatibility of most PTA agents and potential long-term toxicity obstruct their clinical translation. Meanwhile, previous PTA studies are limited to surface tumors because of insufficient light penetration depth of near-infrared (NIR) light for deep abdominal tumors. Therefore, minimally invasive PTA combined with biocompatible agents may pave a promising way to treat deep orthotopic hepatocellular carcinoma (HCC). Herein, a multifunctional agent based on superparamagnetic iron oxide (SPIO) and new indocyanine green (IR820) was constructed with good biocompatibility. Outstanding fluorescence, photoacoustic and magnetic resonance imaging capabilities were observed in vitro. Additionally, in vivo results indicated that early-stage HCC (diameter less than 2 mm) could be effectively detected by this agent. Furthermore, for the first time, we developed minimally invasive laparoscopic-assisted photothermal ablation (L-A PTA) method coupled with this agent to completely ablate orthotopic HCC in nude mice model, neither recurrences nor obvious side effects were observed during the experiments. Remarkable shrinkage of primary tumor and disappearance of intrahepatic metastasis were also observed. In summary, minimally invasive L-A PTA is an effective preoperative neoadjuvant treatment for HCC.
Collapse
|
Research Support, Non-U.S. Gov't |
5 |
28 |
7
|
Zhao H, Wang J, Li X, Li Y, Li C, Wang X, Wang J, Guan S, Xu Y, Deng G, Chen Y, Lu J, Liu X. A biocompatible theranostic agent based on stable bismuth nanoparticles for X-ray computed tomography/magnetic resonance imaging-guided enhanced chemo/photothermal/chemodynamic therapy for tumours. J Colloid Interface Sci 2021; 604:80-90. [PMID: 34265694 DOI: 10.1016/j.jcis.2021.06.174] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 11/17/2022]
Abstract
Cancer is a leading cause of death worldwide and seriously threatens the health of humans. The current clinical treatments for cancer are not efficient and always lead to significant side effects. Herein, a biocompatible and powerful theranostic agent (Bi@mSiO2@MnO2/DOX) is fabricated using a facile stepwise reaction method. The Bi nanoparticles (NPs) are coated by mesoporous silica to protect the Bi NPs from oxidation, which guarantees the stable photothermal effect of the Bi NPs. When the Bi@mSiO2@MnO2/DOX nanocomposites (NCs) accumulate in the tumour site, hyperthermia is generated by Bi NPs under near-infrared (NIR) light irradiation for photothermal therapy (PTT), and the generated heat triggers the release of DOX for chemotherapy in the tumour. In addition, the MnO2 of the NCs responsively catalyses endogenous H2O2 to generate O2, raising the oxygen level to enhance the effect of chemotherapy in the tumour microenvironment (TME), and consumes glutathione (GSH) to produce Mn2+ for magnetic resonance (MR) imaging. Under acidic TME conditions, H2O2 and Mn2+ also produce toxic hydroxyl radical (·OH) for chemodynamic therapy (CDT). Furthermore, the Bi NPs can also be used as excellent contrast agents for X-ray computed tomography (CT) imaging of tumours with a high CT value (6.865 HU mM-1). The Bi@mSiO2@MnO2/DOX NCs exhibit a powerful theranostic performance for CT/MR imaging-guided enhanced PTT/CDT/chemotherapy, which opens a new prospect to rationally design theranostic agents for tumour imaging.
Collapse
|
Journal Article |
4 |
25 |
8
|
Yan WC, Chua QW, Ong XJ, Sharma VK, Tong YW, Wang CH. Fabrication of ultrasound-responsive microbubbles via coaxial electrohydrodynamic atomization for triggered release of tPA. J Colloid Interface Sci 2017; 501:282-293. [PMID: 28460221 DOI: 10.1016/j.jcis.2017.04.073] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 04/21/2017] [Accepted: 04/23/2017] [Indexed: 01/16/2023]
Abstract
A single-step fabrication method, coaxial electrohydrodynamic atomization (CEHDA), was developed to synthesize drug-loaded microbubbles (MBs) for combination treatment of ischemic stroke. The bioactivity of therapeutic agent (tPA, tissue plasminogen activator) after preparation was evaluated, showing that CEHDA could be very promising method for producing MBs with therapeutic functions. The bubble performance and tPA release profiles were also examined by exposing the bubbles to 2MHz ultrasound of various intensities. The results showed that the mean diameter of tPA-loaded MBs was found to fluctuate about its original diameter when exposed to ultrasound and higher intensity ultrasound was more effective in triggering the burst of CEHDA MBs. High ultrasound-triggered bubble disintegration effectiveness in a short period (first 5min) fits well with the requirement of short ultrasound exposure time for human brain. Moreover, a numerical model was also applied to investigate the stability of the fabricated MBs in the bloodstream. It was found that MB dissolution time increased with initial radius, decreased with initial surface tension and increased with initial shell resistance but it was barely affected by the average excessive bloodstream pressure.
Collapse
|
Journal Article |
8 |
23 |
9
|
Khaniabadi PM, Shahbazi-Gahrouei D, Aziz AA, Dheyab MA, Khaniabadi BM, Mehrdel B, Jameel MS. Trastuzumab conjugated porphyrin-superparamagnetic iron oxide nanoparticle: A potential PTT-MRI bimodal agent for herceptin positive breast cancer. Photodiagnosis Photodyn Ther 2020; 31:101896. [PMID: 32585402 DOI: 10.1016/j.pdpdt.2020.101896] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/07/2020] [Accepted: 06/19/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Theranostic agents can combine photosensitizers and contrast agents into a single unit for photothermal therapy (PTT) and magnetic resonance imaging (MRI). The possibility of treating and diagnosing malignant cancers without any ionizing radiation could become an option. This study investigates the theranostic potential of Fe3O4 nanoparticles (IONs) for the diagnosis and treatment of cancer by developing a single integrated nanoprobe. METHODS Oleylamin-coated IONs (ION-Ol) were synthesized and surface of the IONs was modified using protoporphyrin (PP) and trastuzumab (TZ) to develop the TZ-conjugated SPION-porphyrin [ION-PP-TZ]. The structure, morphology, size, and cytotoxicity of all samples were investigated using Fourier-transform infrared spectroscopy (FT-IR), Transmission electron microscopy (TEM), X-ray powder diffraction (XRD), WST-1 assay, respectively. In addition to MRI and in vitro laser irradiation (808 nm, 200 mW) to determine the r2 values and photothermal ablation. RESULTS The sizes of monodispersed nanoparticles were measured in rang 5.74-7.17 nm. No cytotoxicity was observed after incubating MCF 7 cells under various Fe concentrations of nanoparticles and theranostic agents. The transverse relaxation time of the protoporphyrin conjugated to IONs (52.32 mM-1s-1) exceeded that of ION-Ol and TZ-conjugated ION-PP. In addition, the in vitro photothermal ablation of ION-PP-TZ revealed a 74 % MCF 7 cell reduction after 10 min of at the highest Fe concentration (1.00 mg Fe/mL). CONCLUSIONS In summary, the water-soluble ION-PP-TZ is a promising bimodal agent for the diagnosis and treatment of human epidermal growth factor receptor 2-positive breast cancer cells using a T2 MRI contrast agent and photothermal therapy.
Collapse
|
Journal Article |
5 |
23 |
10
|
Lu ST, Xu D, Liao RF, Luo JZ, Liu YH, Qi ZH, Zhang CJ, Ye NL, Wu B, Xu HB. Single-Component Bismuth Nanoparticles as a Theranostic Agent for Multimodal Imaging-Guided Glioma Therapy. Comput Struct Biotechnol J 2019; 17:619-627. [PMID: 31193098 PMCID: PMC6517535 DOI: 10.1016/j.csbj.2019.04.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/07/2019] [Accepted: 04/10/2019] [Indexed: 11/30/2022] Open
Abstract
Single-component nanomaterials such as bismuth (Bi) based on nanoparticles (NPs) intrinsically having both diagnostic and therapeutic capabilities are widely needed in biomedical fields. However, their design and fabrication still face enormous challenges. Here, a kind of pure Bi NPs with ultrahigh X-ray attenuation coeffcient was developed and evaluated as a simple but powerful theranostic nanomaterals and potent light-to-heat conversion efficiency for photoacuostic imaging (PAI)/photothermal therapy (PTT) in this study. The prepared pure Bi NPs showed excellent photothermal performance and the temperature of NPs solution (1 mg/mL) increased to 70 °C under near-infrared light irradiation within 4 min. The pure Bi NPs showed obvious enhancement effect both in X-ray computed tomography (CT) and PA imaging modalities in vivo. In addition, the glioma growth was efficiently suppressed by the pure Bi NPs after 808 nm laser irradiation, while maintained the biosafety and low toxicity. Thus, it is notable that this type of Bi nanomaterial has great potential in multi-imaging guided cancer treatment.
Collapse
|
research-article |
6 |
18 |
11
|
Mirković M, Radović M, Stanković D, Milanović Z, Janković D, Matović M, Jeremić M, Antić B, Vranješ-Đurić S. 99mTc-bisphosphonate-coated magnetic nanoparticles as potential theranostic nanoagent. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 102:124-133. [PMID: 31146983 DOI: 10.1016/j.msec.2019.04.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 04/01/2019] [Accepted: 04/12/2019] [Indexed: 12/20/2022]
Abstract
Novel theranostic nanoplatform is expected to integrate imaging for guiding and monitoring of the tumor therapy with great therapeutic efficacy and fewer side effects. Here we describe the preparation of a multifunctional 99mTc-bisphosphonate-coated magnetic nanoparticles (MNPs) based on Fe3O4 and coated with two hydrophilic bisphosphonate ligands, i.e., methylene diphosphonate (MDP) and 1-hydroxyethane-1,1- diphosphonate (HEDP). The presence of the bisphosphonates on the MNPs surface, enabled their biocompatibility, colloidal stability and successful binding of the radionuclide. The morphology, size, structure, surface charge and magnetic properties of obtained bisphosphonate-coated Fe3O4 MNPs were characterized by transmission electron microscopy, X-ray powder diffraction, dynamic light scattering, laser Doppler electrophoresis, Fourier transform infrared spectroscopy and vibrating sample magnetometer. The specific power absorption values for Fe3O4-MDP and Fe3O4-HEDP were 113 W/g and 141 W/g, respectively, indicated their heating ability under applied magnetic field. Coated MNPs were radiolabeled with 99mTc using stannous chloride as the reducing agent in a reproducible high yield (95% for Fe3O4-MDP and 97% for Fe3O4-HEDP MNPs) and were remained stable in saline and human serum for 24 h. Ex vivo biodistribution studies presented significant liver and spleen uptake in healthy Wistar rats after intravenous administration at all examined time points due to the colloidal nature of both 99mTc-MNPs. Results of scintigraphy studies are in accordance with ex vivo biodistribution studies, demonstrating high in vivo stability of radiolabeled MNPs and therefore results of both methods were proved as accurate information on the biodistribution profile of investigated MNPs. Overall, in vitro and in vivo stability as well as heating ability, indicate that biocompatible radiolabeled bisphosphonate magnetic nanoparticles exhibit promising potential as a theranostic nanoagent.
Collapse
|
Journal Article |
6 |
18 |
12
|
Dheyab MA, Aziz AA, Khaniabadi PM, Jameel MS. Potential of a sonochemical approach to generate MRI-PPT theranostic agents for breast cancer. Photodiagnosis Photodyn Ther 2021; 33:102177. [PMID: 33429101 DOI: 10.1016/j.pdpdt.2021.102177] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/22/2020] [Accepted: 12/31/2020] [Indexed: 01/25/2023]
Abstract
The production of nanomaterials integrating diagnostic and therapeutic roles within one nanoplatform is important for medical applications. Such theranostics nanoplatforms could provide information on imaging, accurate diagnosis and, at the same time, could eradicate cancer cells. Fe3O4@Au core@shell nanoparticles (Fe3O4@AuNPs) have gained broad attention due to their unique innovations in magnetic resonance imaging (MRI) and photothermal therapy (PTT). Seed-mediated growth procedures were used to produce the Fe3O4@AuNPs. In these processes, complicated surface modifications, resulted in unsatisfactory properties. This work used the ability of the sonochemical approach to synthesize highly efficient theranostics agent Fe3O4@AuNPs with a size of approximately 22 nm in 5 min. The inner core of Fe3O4 acts as an MRI agent, whereas the photothermal effect stands accomplished by near-infrared absorption of the gold shell (Au shell), which results in the eradication of cancer cells. We have shown that Fe3O4@AuNPs have great biocompatibility and no major cytotoxicity has been identified. Relaxivity value (r2) of synthesized Fe3O4@Au NPs, measured at 233 mM-1s-1, is significantly higher than those reported previously. The as-synthesized NPs have shown substantial photothermal ablation ability on MCF-7 in vitro under near-infrared laser irradiation. Consequently, Fe3O4@AuNPs synthesized in this study have great potential as an ideal candidate for MR imaging and PTT.
Collapse
|
Journal Article |
4 |
17 |
13
|
Tatar AS, Jurj A, Tomuleasa C, Florea A, Berindan-Neagoe I, Cialla-May D, Popp J, Astilean S, Boca S. CD19-targeted, Raman tagged gold nanourchins as theranostic agents against acute lymphoblastic leukemia. Colloids Surf B Biointerfaces 2019; 184:110478. [PMID: 31541890 DOI: 10.1016/j.colsurfb.2019.110478] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/15/2019] [Accepted: 08/29/2019] [Indexed: 10/26/2022]
Abstract
The incidence of Acute Lymphoblastic Leukemia (ALL) is increasing globally, and it is being clinically addressed by chemotherapy, followed by immunotherapy and stem cell transplantation, all with potential life-threatening toxicities. In the need for more effective therapeutics, newly developed disease-targeted nanocompounds can thus hold real potential. In this paper, we propose a novel nanoparticle-based immunotherapeutic agent against ALL, consisting of antiCD19 antibody-conjugated, polyethylene glycol (PEG)-biocompatibilized, and Nile Blue (NB) Raman reporter-tagged gold nanoparticles of urchin-like shape (GNUs), that have a plasmonic response in the Near Infrared (NIR) spectral range. Transmission electron microscopy (TEM) images of particle-incubated CD19-positive (CD19(+)) CCRF-SB cells show that the antiCD19-PEG-NB-GNU nanocomplex is able to recognize the CD19 B-cell-specific antigen, which is a prerequisite for targeted therapy. The therapeutic effect of the particles is confirmed by cell counting, combined with cell cycle analysis by flow cytometry and MTS assay, which additionally offer insights into their mechanisms of action. Specifically, antiCD19-PEG-NB-GNUs proved superior cytotoxic effect against CCRF-SB cells when compared with the free antibody, by reducing the overall viability below 18% after 7 days treatment at a particle-bound antibody concentration of 0.17 ng/μl. Moreover, by combining their remarkable plasmonic properties with the possibility of Raman tagging, the proposed nanoparticles can also serve as spectroscopic imaging agents inside living cells, which validates their theranostic potential in the field of hematological oncology.
Collapse
|
Journal Article |
6 |
11 |
14
|
A ROS-responsive, self-immolative and self-reporting hydrogen sulfide donor with multiple biological activities for the treatment of myocardial infarction. Bioact Mater 2021; 9:168-182. [PMID: 34820564 PMCID: PMC8586025 DOI: 10.1016/j.bioactmat.2021.07.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/22/2021] [Accepted: 07/09/2021] [Indexed: 12/14/2022] Open
Abstract
Myocardial infarction (MI), as one of the leading causes of global death, urgently needs effective therapies. Recently, hydrogen sulfide (H2S) has been regarded as a promising therapeutic agent for MI, while its spatiotemporally controlled delivery remains a major issue limiting clinical translation. To address this limitation, we designed and synthesized a novel H2S donor (HSD-R) that can produce H2S and emit fluorescence in response to reactive oxygen species (ROS) highly expressed at diseased sites. HSD-R can specifically target mitochondria and provide red fluorescence to visualize and quantify H2S release in vitro and in vivo. Therapeutically, HSD-R significantly promoted the reconstruction of cardiac structure and function in a rat MI model. Mechanistically, myocardial protection is achieved by reducing cardiomyocyte apoptosis, attenuating local inflammation, and promoting angiogenesis. Furthermore, inhibition of typical pro-apoptotic genes (Bid, Apaf-1, and p53) played an important role in the anti-apoptotic effect of HSD-R to achieve cardioprotection, which were identified as new therapeutic targets of H2S against myocardial ischemia injury. This ROS-responsive, self-immolative, and fluorescent H2S donor can serve as a new theranostic agent for MI and other ischemic diseases.
A reactive oxygen species-responsive and self-reporting H2S donor (HSD-R) is developed for controlled H2S delivery. HSD-R shows desirable fluorescence for imaging H2S release upon triggering by reactive oxygen species. HSD-R displays significant cardioprotective effects in rats. HSD-R exhibits multiple biological activities including anti-apoptotic, anti-inflammatory, and pro-angiogenic effects. Anti-apoptotic activity of HSD-R is due to inhibiting the expression of several pro-apoptotic factors.
Collapse
|
|
4 |
9 |
15
|
Tandale P, Choudhary N, Singh J, Sharma A, Shukla A, Sriram P, Soni U, Singla N, Barnwal RP, Singh G, Kaur IP, Suttee A. Fluorescent quantum dots: An insight on synthesis and potential biological application as drug carrier in cancer. Biochem Biophys Rep 2021; 26:100962. [PMID: 33763604 PMCID: PMC7973288 DOI: 10.1016/j.bbrep.2021.100962] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 02/11/2021] [Accepted: 02/22/2021] [Indexed: 02/01/2023] Open
Abstract
Quantum dots (QDs) are nanocrystals of semiconducting material possessing quantum mechanical characteristics with capability to get conjugated with drug moieties. The particle size of QDs varies from 2 to 10 nm and can radiate a wide range of colours depending upon their size. Their wide and diverse usage of QDs across the world is due to their adaptable properties like large quantum yield, photostability, and adjustable emission spectrum. QDs are nanomaterials with inherent electrical characteristics that can be used as drug carrier vehicle and as a diagnostic in the field of nanomedicine. Scientists from various fields are aggressively working for the development of single platform that can sense, can produce a microscopic image and even be used to deliver a therapeutic agent. QDs are the fluorescent nano dots with which the possibilities of the drug delivery to a targeted site and its biomedical imaging can be explored. This review is mainly focused on the different process of synthesis of QDs, their application especially in the areas of malignancies and as a theranostic tool. The attempt is to consolidate the data available for the use of QDs in the biomedical applications.
QDs are nonmaterial's that can be used for drug delivery, imaging and diagnostic tool in the field of nanomedicine. The various approaches to synthesize the QDs were explored. QDs are accepted in the treatment strategies due to their biocompatibility with human physiology. QDs posses' several biomedical application particularly in the area of cancer theranostics. Fluorescents dots (QDs) can illuminate the complicated terrain of oncology sciences, novel biomarkers and a patient compliant treatment regimens.
Collapse
|
Review |
4 |
8 |
16
|
Rizvi SFA, Shahid S, Mu S, Zhang H. Hybridization of tumor homing and mitochondria-targeting peptide domains to design novel dual-imaging self-assembled peptide nanoparticles for theranostic applications. Drug Deliv Transl Res 2021; 12:1774-1785. [PMID: 34535874 DOI: 10.1007/s13346-021-01066-6] [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] [Accepted: 09/09/2021] [Indexed: 01/05/2023]
Abstract
A novel hybridized dual-targeting peptide-based nanoprobe was successfully designed by using the cyclic heptapeptide. This peptide has Arg-Gly-Asp-Lys-Leu-Ala-Lys sequence, in which the RGD homing motif and KALK mitochondria-targeting motif were linked via amide bond. The designed peptide probe was further modified through covalent linkage to induce dual-imaging functionality, and self-assembled to form spherical nanoparticles. The novel Cy5.5-SAPD-99mTc nanoparticles were tested for in vitro cytotoxicity, cellular uptake, and apoptosis-inducing functionalities. The cellular internalization, enhanced cytotoxicity and selective receptor binding capabilities against U87MG cells, excellent dual-imaging potential, improved apoptosis-inducing feature by damaging mitochondria, and in vivo preclinical investigations suggested that our newly designed novel hybridized peptide-based dual-imaging nanoparticles may serve as an admirable theranostic probe to treat brain tumor glioblastoma multiforme. This study describes the development of dual-targeting self-assembled peptide nanoparticles followed by modifications using NIRF dye and radiolabeled with 99mTc for dual-imaging and enhanced therapeutic efficacy against brain tumor.
Collapse
|
|
4 |
6 |
17
|
Yang H, Liu Z, Li X, Zhang Z, Chen D, Lian H. Artesunate-Loaded and Near-Infrared Dye-Conjugated Albumin Nanoparticles as High-Efficiency Tumor-Targeted Photo-Chemo Theranostic Agent. NANOSCALE RESEARCH LETTERS 2018; 13:319. [PMID: 30311065 PMCID: PMC6181830 DOI: 10.1186/s11671-018-2700-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 08/30/2018] [Indexed: 05/20/2023]
Abstract
Herein, a tumor-targeted multifunctional theranostic agent was synthetized using a facile method, combining four clinically approved materials: artesunate (Arte), human serum albumin (HSA), folic acid (FA), and indocyanine green (ICG). The obtained nanocomposites (FA-IHA NPs) showed an excellent photo- and physiological stability. The ICG in the FA-IHA NPs was used not only for near infrared (NIR) fluorescence imaging, but also for photothermal and photodynamic (PTT-PDT) therapy under a single NIR irradiation. In addition, the NIR irradiation (808 nm, 1 W/cm2) could trigger Arte release that showed enhanced chemotherapeutic effect. Through fluorescence imaging, the cell uptake and tumor accumulation of FA-IHA NPs were observed in vitro and in vivo, analyzed by confocal microscopy and NIR fluorescence imaging in tumor xenograft mice. Based on the diagnostic results, FA-IHA NPs at 24 h post injection and combined with NIR irradiation (808 nm, 1 W/cm2) could efficiently suppress tumor growth through a photo-chemo combination therapy, with no tumor recurrence in vitro and in vivo. The obtained results suggested that FA-IHA NPs are promising photo-chemo theranostic agents for future clinical translation.
Collapse
|
brief-report |
7 |
6 |
18
|
Zhang YJ, Li BA, Li ZY, Xia N, Yu HY, Zhang YZ. Synthesis and characterization of Tamoxifen citrate modified reduced graphene oxide nano sheets for breast cancer therapy. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2017; 180:68-71. [PMID: 29413703 DOI: 10.1016/j.jphotobiol.2017.12.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 12/14/2017] [Accepted: 12/15/2017] [Indexed: 10/18/2022]
Abstract
Theranostic agents are of immense consideration in the current generation nanomedicine. In this study, we have developed a facile approach for the fabrication of Tamoxifen citrate modified nanosized reduced graphene oxide (nano-rGO) with more stability and low cytotoxicity. The prepared nano-rGO sheets were characterized using HR-TEM and AFM imaging techniques. Further, the cytotoxicity was assessed using MTT assay on female BALB/c nude mice MCF-7 cell lines. In addition, by means of continuous-wave near-infrared laser, cancer cells in vivo were significantly ablated because of the photothermal effect stimulated by tamoxifen modified nano-rGO. These results indicated that the prepared tamoxifen modified nano-rGO has the ability to apply in the photothermal therapy of breast cancers. Consequently, further exploration of photothermal therapeutics is desirable for the synthesis of novel nano materials with additional functionalities.
Collapse
|
Journal Article |
8 |
5 |
19
|
Auria-Luna F, Marqués-López E, Romanos E, Fernández-Moreira V, Gimeno MC, Marzo I, Herrera RP. Novel ureido-dihydropyridine scaffolds as theranostic agents. Bioorg Chem 2020; 105:104364. [PMID: 33113409 DOI: 10.1016/j.bioorg.2020.104364] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/25/2020] [Accepted: 10/07/2020] [Indexed: 12/13/2022]
Abstract
In this work, the synthesis of interesting urea derivatives 5 based on 1,4-dihydropyridines 3 is described for the first time. Considering that both families exhibit potential as drugs to treat various diseases, their activity as anticancer agents has been evaluated in HeLa (cervix), Jurkat (leukaemia) and A549 (lung) cancer cell lines as well as on healthy mice in vivo. In general, whereas 1,4-dihydropyridines show a moderate cytotoxic activity, their urea analogues cause an extraordinary increase in their antiproliferative activity, specially towards HeLa cells. Because of the chiral nature of these compounds, enantiomerically enriched samples were also tested, showing different cytotoxic activity than the racemic mixture. Although the reason is not clear, it could be caused by a complex amalgam of physical and chemical contributions. The studied compounds also exhibit luminescent properties, which allow performing a biodistribution study in cancer cells. They have emission maxima between 420 and 471 nm, being the urea derivatives in general red shifted. Emission quenching was observed for those compounds containing a nitro group (3e,f and 5e,f). Fluorescence microscopy showed that 1,4-dihydropyridines 3a and 3g localised in the lysosomes, in contrast to the urea derivatives 5h that accumulated in the cell membrane. This different distribution could be key to explain the differences found in the cytotoxic activity and in the mechanism of action. Interestingly, a preliminary in vivo study regarding the acute toxicity of some of these compounds on healthy mice has been conducted, using a concentration up to 7200 times higher than the corresponding IC50 value. No downgrade in the welfare of the tested mice was observed, which could support their use in preclinical tumour models.
Collapse
|
Research Support, Non-U.S. Gov't |
5 |
2 |
20
|
Kou X, Shi X, Pang Z, Yang A, Shen R, Zhao L. A Review on the Natural Components Applied as Lead Compounds for Potential Multi-target Anti-AD Theranostic Agents. Curr Med Chem 2023:CMC-EPUB-129088. [PMID: 36698238 DOI: 10.2174/0929867330666230125153027] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/17/2022] [Accepted: 11/24/2022] [Indexed: 01/26/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that seriously affects the health and quality of life of the elderly. Its pathogenesis is very complex and there is still a lack of effective clinical drugs to treat or control the development of AD. Studies have shown that β-amyloid (Aβ) deposition, tau protein hyperphosphorylation, reduced levels of brain cholinergic transmitters, and oxidative stress are the main causes of AD. Furthermore, recent studies showed that metal dyshomeostasis could relate to all the above pathogenesis of AD and was a key factor in the development of AD. Natural compounds and their derivatives have multi-target therapeutic effects on AD, and they also have the advantages of low toxicity, and low cost, which are important directions for anti-AD drugs. Meanwhile, early detection may play an important role in in preventing the development of AD. The concept of "theranostic agent" combining molecular imaging probes and therapeutic drugs has emerged in recent years. Fluorescence imaging has been widely studied and applied because of its non-invasive, high resolution, high sensitivity, rapid imaging, and low cost. However, at present, most of the research methods in this field use individual therapeutic or diagnostic reagents, which is not conducive to exploring the optimal treatment time window and drug efficacy. Therefore, this work reviewed the natural compounds and their derivatives which all have been studied for both the in vitro and in vivo therapeutic and diagnostic anti-AD activities. At last, structure and activity relationship (SAR) was discussed and potential AD theranostic natural agents were put forwarded to provide a more detailed theoretical basis for the further development of drugs with diagnostic and therapeutic effects in AD.
Collapse
|
|
2 |
2 |
21
|
A Rational design of multi-functional nanoplatform: Fluorescent-based "off-on" theranostic gold nanoparticles modified with D-α-Tocopherol succinate. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2021; 222:112261. [PMID: 34330081 DOI: 10.1016/j.jphotobiol.2021.112261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 11/23/2022]
Abstract
It is crucial to develop nanocarrier systems to detect and treat drug-resistant micro tumors to prevent recurrence and/or metastasis of cancer. Due to their exceptional features such as biocompatibility, easy surface modification, serving as imaging and therapeutic agent, gold nanoparticles (AuNPs) draw attention as theranostic agents. It is beneficial to combine AuNPs with a second imaging and/or treatment modality such as photodynamic therapy (PDT). PDT is a non-mutagenic treatment approach in which photosensitizer is activated with light, generating reactive oxygen species and/or free radicals to destroy tumor cells. With the aim of developing "off-on" theranostic system, citrate stabilized spherical 13 nm AuNPs were densely coated with polyethylene glycol (PEG). To advance the theranostic feature of PEGylated AuNPs, they were further functionalized with FDA-Approved photosensitizer, Verteporfin (BPD-MA). Due to static quenching between BPD-MA and AuNPs as well as in between nearby BPD-MA molecules, the fluorescence of the ground state complex is quenched and the system is in "off" state. When BPD-MA molecules are cleaved from the AuNPs surface and diffuse away, fluorescence is recovered. Consequently, the system switches to the "on" state. Among the various mole ratios of BPD-MA carrying conjugates prepared, the most promising candidate was selected based on stability, quenching factor, and fluorescence recovery rate. The conjugate was further decorated with D-α-Tocopherol succinate (VitES) to increase the therapeutic efficacy of the theranostic agent via enhancing cellular uptake. Our results showed that it was possible to achieve as high as 80 times fluorescence quenching when the system was "off". As the system switched from "off" to "on" state, 51% of the fluorescence was recovered. When BPD-MA was immobilized on the PEGylated AuNPs, the phototoxic effect of BPD-MA increased twice against the MCF-7 cell line. Moreover, the developed system showed four times more phototoxicity than BPD-MA alone after it was decorated with VitES. Since the developed system is capable of dual imaging (computed tomography and fluorescence) and dual treatment (PDT and hyperthermia), it potentially offers superior imaging and therapy options for various types of in vitro/in vivo applications.
Collapse
|
Journal Article |
4 |
2 |
22
|
Kou X, Li X, Hu C, Liu J, Chen Y, Zhang Y, Yang A, Shen R. Multifunctional fluorescence sensor as a potential theranostic agent against Alzheimer's disease. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 267:120587. [PMID: 34782268 DOI: 10.1016/j.saa.2021.120587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 10/29/2021] [Accepted: 11/03/2021] [Indexed: 06/13/2023]
Abstract
Metal ions play an important role in the pathogenesis of Alzheimer's disease (AD). Metal dyshomeostasis, β-amyloid (Aβ) accumulation and oxidative stress, etc. are related to metal ions. So, metal therapeutics has aroused increasingly more attention, especially the research of metal-involved theranostic agents. In this work, a highly selective and sensitive multifunctional fluorescence sensor 1 with a naphthol unit based on photoinduced electron transfer (PET) and excited state proton transfer (ESPT) mechanism was synthesized, and its synergistic biological effects on regulating metal dyshomeostasis, modulating Aβ accumulation and scavenging reactive oxygen species (ROS) was evaluated. The results demonstrated that 1 exhibited significant fluorescence enhancement towards Al3+ (the limit was as low as 0.01 ppm), superior chelating abilities with metal ions, even better modulation effect of Cu2+-induced Aβ1-42 accumulation than curcumin, good elimination effect of ROS, clear fluorescence image in living cells, low cytotoxic and appropriate blood brain barrier (BBB) permeability. Overall, these findings revealed that 1 could be used as a potential theranostic agent against AD for further research.
Collapse
|
|
3 |
1 |
23
|
Gowsalya K, Rithisa B, Haldorai Y, Shanthi K, Vivek R. Engineered photonic near-infrared light activated photothermal theranostic nanovaccine induced targeted remodeling of tumor microenvironment. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 57:102738. [PMID: 38341011 DOI: 10.1016/j.nano.2024.102738] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/10/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
Tumor recurrence, which happens as a result of persisting tumor cells and minor lesions after treatments like surgery and chemotherapy, is a major problem in oncology. Herein, a strategy to combat this issue by utilize a theranostic nanovaccine composed of photonic HCuS. This nanovaccine aims to eradicate cancer cells and their traces while also preventing tumor recurrence via optimizing the photothermal immune impact. Successful membrane targeting allows for the introduction of new therapeutic agents into the tumor cells. Together with co-encapsulated Toll-Like Receptors (TLR7/8) agonist R848 for activating T cells and maturing DCs, the combined effects of HCuS and ICG function as photothermal agents that generate heat in the presence of NIR light. Photothermal-mediated immunotherapy with therapeutic modalities proved successful in killing tumor cells. By activating the immune system, this new photonic nanovaccine greatly increases immunogenic cell death (ICD), kills tumor cells, and prevents their recurrence.
Collapse
|
|
1 |
1 |
24
|
Gupta I, Su X, Jonnalagadda US, Das D, Pramanik M, Kwan JJ. Investigating the Acoustic Response and Contrast Enhancement of Drug-Loadable PLGA Microparticles with Various Shapes and Morphologies. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:1844-1856. [PMID: 33810888 DOI: 10.1016/j.ultrasmedbio.2021.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/07/2021] [Accepted: 02/16/2021] [Indexed: 06/12/2023]
Abstract
Polymer nanoparticles and microparticles have been used primarily for drug delivery. There is now growing interest in further developing polymer-based solid cavitation agents to also enhance ultrasound imaging. We previously reported on a facile method to produce hollow poly(lactic-co-glycolic acid) (PLGA) microparticles with different diameters and degrees of porosity. Here, we investigate the cavitation response from these PLGA microparticles with both therapeutic and diagnostic ultrasound transducers. Interestingly, all formulations exhibited stable cavitation; larger porous and multicavity particles also provided inertial cavitation at elevated acoustic pressure amplitudes. These larger particles also achieved contrast enhancement comparable to that of commercially available ultrasound contrast agents, with a maximum recorded contrast-to-tissue ratio of 28 dB. Therefore, we found that multicavity PLGA microparticles respond to both therapeutic and diagnostic ultrasound and may be applied as a theranostic agent.
Collapse
|
|
4 |
|
25
|
Lu H, Zeng H, Wei W, Chen Y, Zhou Z, Ning X, Hu P. A fluorogenic ROS-triggered hydrogen sulfide donor for alleviating cerebral ischemia-reperfusion injury. Theranostics 2024; 14:7589-7603. [PMID: 39659579 PMCID: PMC11626942 DOI: 10.7150/thno.100930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/03/2024] [Indexed: 12/12/2024] Open
Abstract
Rationale: Cerebral ischemia-reperfusion injury is a severe neurovascular disease that urgently requires effective therapeutic interventions. Recently, hydrogen sulfide (H2S) has garnered significant attention as a potential treatment for stroke; however, the precise and targeted delivery of H2S remains a considerable challenge for its clinical application. Methods: We have developed HSDF-NH2, a novel H2S donor characterized by high selectivity, self-reporting capabilities, and the ability to penetrate the blood-brain barrier (BBB). Results: HSDF-NH2 effectively scavenges reactive oxygen species (ROS) while generating H2S, with emitted fluorescence facilitating the visualization and quantification of H2S release. This compound has demonstrated protective effects against cerebral ischemia-reperfusion (I/R) injury and contributes to the reconstruction of brain structure and function in a rat stroke model (tMCAO/R). Conclusion: As a ROS-responsive, self-reporting, and fluorescent H2S donor, HSDF-NH2 holds considerable promise for the treatment of ischemic diseases beyond stroke.
Collapse
|
research-article |
1 |
|