Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.915
Filter
1.
Nat Commun ; 15(1): 6610, 2024 Aug 04.
Article in English | MEDLINE | ID: mdl-39098904

ABSTRACT

There is no effective and noninvasive solution for thrombolysis because the mechanism by which certain thrombi become tissue plasminogen activator (tPA)-resistant remains obscure. Endovascular thrombectomy is the last option for these tPA-resistant thrombi, thus a new noninvasive strategy is urgently needed. Through an examination of thrombi retrieved from stroke patients, we found that neutrophil extracellular traps (NETs), ε-(γ-glutamyl) lysine isopeptide bonds and fibrin scaffolds jointly comprise the key chain in tPA resistance. A theranostic platform is designed to combine sonodynamic and mechanical thrombolysis under the guidance of ultrasonic imaging. Breakdown of the key chain leads to a recanalization rate of more than 90% in male rat tPA-resistant occlusion model. Vascular reconstruction is observed one month after recanalization, during which there was no thrombosis recurrence. The system also demonstrates noninvasive theranostic capabilities in managing pigs' long thrombi (>8 mm) and in revascularizing thrombosis-susceptible tissue-engineered vascular grafts, indicating its potential for clinical application. Overall, this noninvasive theranostic platform provides a new strategy for treating tPA-resistant thrombi.


Subject(s)
Thrombolytic Therapy , Thrombosis , Tissue Plasminogen Activator , Animals , Tissue Plasminogen Activator/therapeutic use , Humans , Thrombosis/diagnostic imaging , Thrombosis/drug therapy , Male , Rats , Thrombolytic Therapy/methods , Extracellular Traps/metabolism , Swine , Fibrinolytic Agents/therapeutic use , Fibrinolytic Agents/pharmacology , Rats, Sprague-Dawley , Disease Models, Animal , Fibrin/metabolism , Theranostic Nanomedicine/methods , Drug Resistance , Stroke/diagnostic imaging , Stroke/therapy , Stroke/drug therapy
2.
Molecules ; 29(15)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39124859

ABSTRACT

Since the discovery of cisplatin's antitumoral activity and its approval as an anticancer drug, significant efforts have been made to enhance its physiological stability and anticancer efficacy and to reduce its side effects. With the rapid development of targeted and personalized therapies, and the promising theranostic approach, platinum drugs have found new opportunities in more sophisticated systems. Theranostic agents combine diagnostic and therapeutic moieties in one scaffold, enabling simultaneous disease monitoring, therapy delivery, response tracking, and treatment efficacy evaluation. In these systems, the platinum core serves as the therapeutic agent, while the functionalized ligand provides diagnostic tools using various imaging techniques. This review aims to highlight the significant role of platinum-based complexes in theranostic applications, and, to the best of our knowledge, this is the first focused contribution on this type of platinum compounds. This review presents a brief introduction to the development of platinum chemotherapeutic drugs, their limitations, and resistance mechanisms. It then describes recent advancements in integrating platinum complexes with diagnostic agents for both tumor treatment and monitoring. The main body is organized into three categories based on imaging techniques: fluorescence, positron emission tomography (PET), single-photon emission computed tomography (SPECT), and magnetic resonance imaging (MRI). Finally, this review outlines promising strategies and future perspectives in this evolving field.


Subject(s)
Antineoplastic Agents , Neoplasms , Theranostic Nanomedicine , Humans , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Neoplasms/drug therapy , Neoplasms/diagnostic imaging , Animals , Positron-Emission Tomography/methods , Magnetic Resonance Imaging , Platinum Compounds/chemistry , Platinum Compounds/therapeutic use , Platinum Compounds/pharmacology , Precision Medicine/methods
4.
Anal Chem ; 96(33): 13557-13565, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39115161

ABSTRACT

Although targeted therapy has revolutionized oncotherapy, engineering a versatile oncotherapy nanoplatform integrating both diagnostics and therapeutics has always been an intractable challenge to overcome the limitations of monotherapy. Herein, a theranostics platform based on DI/MP-MB has successfully realized the fluorescence detection of disease marker miR-21 and the gene/photothermal/chemo triple synergetic cancer therapy, which can trace the tumor through photothermal and fluorescence dual-mode imaging and overcome the limitations of monotherapy to improve the treatment efficiency of tumors. DI/MP-MB was prepared by magnetic mesoporous silicon nanoparticles (M-MSNs) loaded with doxorubicin (Dox) and new indocyanine green (IR820), and subsequently coating polydopamine as a "gatekeeper", followed by the surface adsorbed with molecular beacons capable of targeting miR-21 for responsive imaging. Under the action of enhanced permeability retention and external magnetic field, DI/MP-MB were targeted and selectively accumulated in the tumor. MiR-21 MB hybridized with miR-21 to form a double strand, which led to the desorption of miR-21 MB from the polydopamine surface and the fluorescence recovery to realize gene silencing and fluorescence imaging for tracking the treatment process. Meanwhile, with the response to the near-infrared irradiation and the tumor's microacid environment, the outer layer polydopamine will decompose, releasing Dox and IR820 to realize chemotherapy and photothermal therapy. Finally, the ability of DI/MP-MB to efficiently suppress tumor growth was comprehensively assessed and validated both in vitro and in vivo. Noteworthily, the excellent anticancer efficiency by the synergistic effect of gene/photothermal/chemo triple therapy of DI/MP-MB makes it an ideal nanoplatform for tumor therapy and imaging.


Subject(s)
Doxorubicin , Indoles , MicroRNAs , Multimodal Imaging , Polymers , Silicon , Theranostic Nanomedicine , Indoles/chemistry , Polymers/chemistry , Silicon/chemistry , Humans , Animals , Doxorubicin/chemistry , Doxorubicin/pharmacology , Mice , Porosity , Indocyanine Green/chemistry , Mice, Nude , Mice, Inbred BALB C , Nanoparticles/chemistry , Cell Line, Tumor , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Optical Imaging , Surface Properties
5.
Mikrochim Acta ; 191(9): 541, 2024 08 16.
Article in English | MEDLINE | ID: mdl-39150483

ABSTRACT

The solid tumors provide a series of biological barriers in cellular microenvironment for designing drug delivery methods based on advanced stimuli-responsive materials. These intertumoral and intratumoral barriers consist of perforated endotheliums, tumor cell crowding, vascularity, lymphatic drainage blocking effect, extracellular matrix (ECM) proteins, hypoxia, and acidosis. Triggering opportunities have been drawn for solid tumor therapies based on single and dual stimuli-responsive drug delivery systems (DDSs) that not only improved drug targeting in deeper sites of the tumor microenvironments, but also facilitated the antitumor drug release efficiency. Single and dual stimuli-responsive materials which are known for their lowest side effects can be categorized in 17 main groups which involve to internal and external stimuli anticancer drug carriers in proportion to microenvironments of targeted solid tumors. Development of such drug carriers can circumvent barriers in clinical trial studies based on their superior capabilities in penetrating into more inaccessible sites of the tumor tissues. In recent designs, key characteristics of these DDSs such as fast response to intracellular and extracellular factors, effective cytotoxicity with minimum side effect, efficient permeability, and rate and location of drug release have been discussed as core concerns of designing paradigms of these materials.


Subject(s)
Antineoplastic Agents , Drug Delivery Systems , Neoplasms , Tumor Microenvironment , Humans , Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Tumor Microenvironment/drug effects , Drug Delivery Systems/methods , Drug Carriers/chemistry , Animals , Theranostic Nanomedicine/methods
6.
Int J Mol Sci ; 25(15)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39126121

ABSTRACT

The burgeoning field of cancer theranostics has witnessed advancements through the development of targeted molecular agents, particularly peptides. These agents exploit the overexpression or mutations of specific receptors, such as the Epidermal Growth Factor receptor (EGFR) and αVß3 integrin, which are pivotal in tumor growth, angiogenesis, and metastasis. Despite the extensive research into and promising outcomes associated with antibody-based therapies, peptides offer a compelling alternative due to their smaller size, ease of modification, and rapid bioavailability, factors which potentially enhance tumor penetration and reduce systemic toxicity. However, the application of peptides in clinical settings has challenges. Their lower binding affinity and rapid clearance from the bloodstream compared to antibodies often limit their therapeutic efficacy and diagnostic accuracy. This overview sets the stage for a comprehensive review of the current research landscape as it relates to EGFR- and integrin αVß3-targeting peptides. We aim to delve into their synthesis, radiolabeling techniques, and preclinical and clinical evaluations, highlighting their potential and limitations in cancer theranostics. This review not only synthesizes the extant literature to outline the advancements in peptide-based agents targeting EGFR and integrin αVß3 but also identifies critical gaps that could inform future research directions. By addressing these gaps, we contribute to the broader discourse on enhancing the diagnostic precision and therapeutic outcomes of cancer treatments.


Subject(s)
ErbB Receptors , Integrin alphaVbeta3 , Neoplasms , Peptides , Radiopharmaceuticals , Humans , Integrin alphaVbeta3/metabolism , ErbB Receptors/metabolism , ErbB Receptors/antagonists & inhibitors , Radiopharmaceuticals/therapeutic use , Radiopharmaceuticals/chemistry , Neoplasms/diagnostic imaging , Neoplasms/therapy , Neoplasms/metabolism , Neoplasms/drug therapy , Peptides/chemistry , Peptides/therapeutic use , Animals , Precision Medicine/methods , Theranostic Nanomedicine/methods
7.
J Nanobiotechnology ; 22(1): 484, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138477

ABSTRACT

BACKGROUND: Inflammatory bowel disease (IBD) is a progressive and debilitating inflammatory disease of the gastrointestinal tract (GIT). Despite recent advances, precise treatment and noninvasive monitoring remain challenging. METHODS: Herein, we developed orally-administered, colitis-targeting and hyaluronic acid (HA)-modified, core-shell curcumin (Cur)- and cerium oxide (CeO2)-loaded nanoprobes (Cur@PC-HA/CeO2 NPs) for computed tomography (CT) imaging-guided treatment and monitoring of IBD in living mice. RESULTS: Following oral administration, high-molecular-weight HA maintains integrity with little absorption in the upper GIT, and then actively accumulates at local colitis sites owing to its colitis-targeting ability, leading to specific CT enhancement lasting for 24 h. The retained NPs are further degraded by hyaluronidase in the colon to release Cur and CeO2, thereby exerting anti-inflammatory and antioxidant effects. Combined with the ability of NPs to regulate intestinal flora, the oral NPs result in substantial relief in symptoms. Following multiple treatments, the gradually decreasing range of the colon with high CT attenuation correlates with the change in the clinical biomarkers, indicating the feasibility of treatment response and remission. CONCLUSION: This study provides a proof-of-concept for the design of a novel theranostic integration strategy for concomitant IBD treatment and the real-time monitoring of treatment responses.


Subject(s)
Cerium , Curcumin , Hyaluronic Acid , Inflammatory Bowel Diseases , Nanoparticles , Theranostic Nanomedicine , Animals , Inflammatory Bowel Diseases/drug therapy , Mice , Cerium/chemistry , Curcumin/pharmacology , Curcumin/chemistry , Curcumin/therapeutic use , Theranostic Nanomedicine/methods , Administration, Oral , Nanoparticles/chemistry , Hyaluronic Acid/chemistry , Hyaluronoglucosaminidase/metabolism , Tomography, X-Ray Computed , Mice, Inbred C57BL , Colon/diagnostic imaging , Colon/pathology , Colon/metabolism , Humans , Colitis/drug therapy
8.
Article in English | MEDLINE | ID: mdl-39140128

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most recalcitrant cancers due to its late diagnosis, poor therapeutic response, and highly heterogeneous microenvironment. Nanotechnology has the potential to overcome some of the challenges to improve diagnostics and tumor-specific drug delivery but they have not been plausibly viable in clinical settings. The review focuses on active targeting strategies to enhance pancreatic tumor-specific uptake for nanoparticles. Additionally, this review highlights using actively targeted liposomes, micelles, gold nanoparticles, silica nanoparticles, and iron oxide nanoparticles to improve pancreatic tumor targeting. Active targeting of nanoparticles toward either differentially expressed receptors or PDAC tumor microenvironment (TME) using peptides, antibodies, small molecules, polysaccharides, and hormones has been presented. We focus on microenvironment-based hallmarks of PDAC and the potential for actively targeted nanoparticles to overcome the challenges presented in PDAC. It describes the use of nanoparticles as contrast agents for improved diagnosis and the delivery of chemotherapeutic agents that target various aspects within the TME of PDAC. Additionally, we review emerging nano-contrast agents detected using imaging-based technologies and the role of nanoparticles in energy-based treatments of PDAC. This article is categorized under: Implantable Materials and Surgical Technologies > Nanoscale Tools and Techniques in Surgery Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Diagnostic Tools > In Vivo Nanodiagnostics and Imaging.


Subject(s)
Nanoparticles , Pancreatic Neoplasms , Theranostic Nanomedicine , Humans , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/drug therapy , Animals , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Tumor Microenvironment , Drug Delivery Systems , Mice , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/diagnostic imaging
9.
Int J Mol Sci ; 25(15)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39125669

ABSTRACT

Advanced breast cancer remains a significant oncological challenge, requiring new approaches to improve clinical outcomes. This study investigated an innovative theranostic agent using the MCM-41-NH2-DTPA-Gd3⁺-MIH nanomaterial, which combined MRI imaging for detection and a novel chemotherapy agent (MIH 2.4Bl) for treatment. The nanomaterial was based on the mesoporous silica type, MCM-41, and was optimized for drug delivery via functionalization with amine groups and conjugation with DTPA and complexation with Gd3+. MRI sensitivity was enhanced by using gadolinium-based contrast agents, which are crucial in identifying early neoplastic lesions. MIH 2.4Bl, with its unique mesoionic structure, allows effective interactions with biomolecules that facilitate its intracellular antitumoral activity. Physicochemical characterization confirmed the nanomaterial synthesis and effective drug incorporation, with 15% of MIH 2.4Bl being adsorbed. Drug release assays indicated that approximately 50% was released within 8 h. MRI phantom studies demonstrated the superior imaging capability of the nanomaterial, with a relaxivity significantly higher than that of the commercial agent Magnevist. In vitro cellular cytotoxicity assays, the effectiveness of the nanomaterial in killing MDA-MB-231 breast cancer cells was demonstrated at an EC50 concentration of 12.6 mg/mL compared to an EC50 concentration of 68.9 mg/mL in normal human mammary epithelial cells (HMECs). In vivo, MRI evaluation in a 4T1 syngeneic mouse model confirmed its efficacy as a contrast agent. This study highlighted the theranostic capabilities of MCM-41-NH2-DTPA-Gd3⁺-MIH and its potential to enhance breast cancer management.


Subject(s)
Breast Neoplasms , Magnetic Resonance Imaging , Nanoparticles , Silicon Dioxide , Theranostic Nanomedicine , Silicon Dioxide/chemistry , Animals , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Female , Theranostic Nanomedicine/methods , Magnetic Resonance Imaging/methods , Mice , Cell Line, Tumor , Nanoparticles/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Contrast Media/chemistry , Gadolinium/chemistry , Porosity , Xenograft Model Antitumor Assays
10.
Acta Biomater ; 184: 397-408, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38960111

ABSTRACT

Photothermal therapy (PTT) has attracted attention as a highly effective non-invasive treatment method. However, the high localized temperatures (>50 °C) required for its treatment will inevitably cause damage to the surrounding normal tissues. Therefore, it is important to develop novel and effective strategies to achieve mild photothermal therapy (mPTT). The overexpression of heat shock proteins (HSPs), a widespread heat stress protein, leads to the generation of heat resistance in cancer cells, which seriously affects the therapeutic effect. Thus, inhibiting the expression of HSPs to reduce the heat resistance of tumor cells is expected to enhance the therapeutic effect of mPTT. Here, we successfully synthesized a fluorescent probe bonded with an amphiphilic polypeptide to a cyanine dye and achieved physical encapsulation of the blocker SB705498 through a self-assembly process. SB705498 promotes transient receptor potential vanilloid member 1 (TRPV1) channel blockade that can inhibit the translocation of the heat shock transcription factor 1 (HSF 1) by blocking the influx of calcium and thus affecting the expression of HSPs, which has the potential to enhance the thermotherapy of cancer under mild conditions. In addition, the nanoparticles enabled NIR-II fluorescence imaging with good stability and high photothermal conversion efficiency (48.10 %). Therefore, this study provides a new strategy for realizing precise mPTT(<45 °C) guided by NIR-II imaging. STATEMENT OF SIGNIFICANCE: Inhibition of overexpression of heat shock proteins (HSPs) in cancer photothermal therapy (PTT) is expected to enhance the therapeutic effect of mild photothermal therapy (mPTT). In this study, we synthesized a fluorescent probe bonded to cyanine dyes with amphiphilic polypeptides and physically wrapped the blocker SB705498 through a self-assembly process. As a transient receptor potential vanillin 1 (TRPV1) channel blocker, SB705498 inhibits heat shock transcription factor 1 (HSF1) translocation by blocking calcium ion influx, thereby improving mPTT efficacy by inhibiting the expression of HSPs. The nanoparticles also enable NIR-II fluorescence imaging with good stability and high photothermal conversion efficiency (48.10 %). Thus, this study provides a new strategy for NIR-II mPTT.


Subject(s)
Infrared Rays , Nanoparticles , Peptides , Photothermal Therapy , TRPV Cation Channels , Nanoparticles/chemistry , TRPV Cation Channels/metabolism , Humans , Peptides/chemistry , Peptides/pharmacology , Theranostic Nanomedicine/methods , Animals , Fluorescent Dyes/chemistry , Heat Shock Transcription Factors/metabolism , Cell Line, Tumor , Mice , Mice, Nude
11.
J Mater Chem B ; 12(31): 7543-7556, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38978513

ABSTRACT

Extracellular clustering of amyloid-ß (Aß) and an impaired autophagy lysosomal pathway (ALP) are the hallmark features in the early stages of incurable Alzheimer's disease (AD). There is a pressing need to find or develop new small molecules for diagnostics and therapeutics for the early stages of AD. Herein, we report a small molecule, namely F-SLCOOH, which can bind and detect Aß1-42, Iowa mutation Aß, Dutch mutation Aß fibrils and oligomers exhibiting enhanced emission with high affinity. Importantly, F-SLCOOH can readily pass through the blood-brain barrier and shows highly selective binding toward the extracellular Aß aggregates in real-time in live animal imaging of a 5XFAD mice model. In addition, a high concentration of F-SLCOOH in both brain and plasma of wildtype mice after intraperitoneal administration was found. The ex vivo confocal imaging of hippocampal brain slices indicated excellent colocalization of F-SLCOOH with Aß positive NU1, 4G8, 6E10 A11 antibodies and THS staining dye, affirming its excellent Aß specificity and targetability. The molecular docking studies have provided insight into the unique and specific binding of F-SLCOOH with various Aß species. Importantly, F-SLCOOH exhibits remarkable anti-fibrillation properties against toxic Aß aggregate formation of Aß1-42, Iowa mutation Aß, and Dutch mutation Aß. F-SLCOOH treatment also exerts high neuroprotective functions and promotes autophagy lysosomal biogenesis in neuronal AD cell models. In summary, the present results suggest that F-SLCOOH is a highly promising theranostic agent for diagnosis and therapeutics of AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Lysosomes , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Mice , Amyloid beta-Peptides/metabolism , Lysosomes/metabolism , Humans , Mutation , Molecular Docking Simulation , Plaque, Amyloid/metabolism , Theranostic Nanomedicine , Mice, Transgenic
12.
Chem Commun (Camb) ; 60(65): 8526-8536, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39039905

ABSTRACT

As we all know, cancer is still a disease that we are struggling against. Although the traditional treatment options are still the mainstream in clinical practice, emerging phototheranostics technologies based on photoacoustic or fluorescence imaging-guided phototherapy also provide a new exploration direction for non-invasive, low-risk and highly efficient cancer treatment. Photosensitizers are the core materials to accomplish this mission. Recently, more attention has been paid to the emerging A-D-A fused-ring photosensitizers. A-D-A fused-ring photosensitizers display strong and wide absorption spectra, high photostability and easy molecular modification. Since this type of photosensitizer was first used for tumor therapy in 2019, its application boundaries are constantly expanding. Therefore, in this feature article, from the perspective of molecular design, we focused on the development of these molecules for application in phototheranostics over the past five years. The effects of tiny structural changes on their photophysical properties are discussed in detail, which provides a way for structural optimization of the subsequent A-D-A photosensitizers.


Subject(s)
Neoplasms , Photosensitizing Agents , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Humans , Neoplasms/drug therapy , Neoplasms/diagnostic imaging , Theranostic Nanomedicine , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Molecular Structure , Photochemotherapy , Phototherapy , Animals
13.
Molecules ; 29(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38999044

ABSTRACT

BACKGROUND: FAP radiopharmaceuticals show promise for cancer diagnosis; however, their limited tumor residency hinders treatment. This study compared two FAPi derivatives, DOTA.SA.FAPi and DOTAGA.(SA.FAPi)2, labeled with gallium-68 and lutetium-177, aiming to determine an optimum combination for creating theranostic pairs. METHODS: The radiotracers were studied for lipophilicity, binding to human serum proteins, and binding to human cancer-associated fibroblasts (CAFs) in vitro, including saturation and internalization/externalization studies. PET/SPECT/CT and biodistribution studies were conducted in PC3 and U87MG xenografts for [68Ga]Ga-DOTA.SA.FAPi and [68Ga]Ga-DOTAGA.(SA.FAPi)2. [177Lu]Lu-DOTA.SA.FAPi and [177Lu]Lu-DOTAGA.(SA.FAPi)2, were evaluated in PC3 xenografts. Biodistribution studies of [68Ga]Ga-DOTA.SA.FAPi were performed in healthy male and female mice. RESULTS: All radiotracers exhibited strong binding to FAP. Their internalization rate was fast while only [177Lu]Lu-DOTAGA.(SA.FAPi)2 was retained longer in CAFs. [68Ga]Ga-DOTAGA.(SA.FAPi)2 and [177Lu]Lu-DOTAGA.(SA.FAPi)2 displayed elevated lipophilicity and affinity for human serum proteins compared to [68Ga]Ga-DOTA.SA.FAPi and [177Lu]Lu-DOTA.SA.FAPi. In vivo studies revealed slower washout of [68Ga]Ga-DOTAGA.(SA.FAPi)2 within 3 h compared to [68Ga]Ga-DOTA.SA.FAPi. The tumor-to-tissue ratios of [68Ga]Ga-DOTAGA.(SA.FAPi)2 versus [68Ga]Ga-DOTA.SA.FAPi did not exhibit any significant differences. [177Lu]Lu-DOTAGA.(SA.FAPi)2 maintained a significant tumor uptake even after 96 h p.i. compared to [177Lu]Lu-DOTA.SA.FAPi. CONCLUSIONS: Dimeric compounds hold promise for therapy, while monomers are better suited for diagnostics. Finding the right combination is essential for effective disease management.


Subject(s)
Endopeptidases , Gallium Radioisotopes , Lutetium , Radioisotopes , Radiopharmaceuticals , Lutetium/chemistry , Humans , Animals , Mice , Tissue Distribution , Radioisotopes/chemistry , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/pharmacology , Gallium Radioisotopes/chemistry , Cell Line, Tumor , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Gelatinases/antagonists & inhibitors , Gelatinases/metabolism , Heterocyclic Compounds, 1-Ring/chemistry , Female , Male , Theranostic Nanomedicine
14.
Biomater Sci ; 12(15): 3805-3825, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38967109

ABSTRACT

Stimulus-responsive polymers have found widespread use in biomedicine due to their ability to alter their own structure in response to various stimuli, including internal factors such as pH, reactive oxygen species (ROS), and enzymes, as well as external factors like light. In the context of atherosclerotic cardiovascular diseases (CVDs), stimulus-response polymers have been extensively employed for the preparation of smart nanocarriers that can deliver therapeutic and diagnostic drugs specifically to inflammatory lesions. Compared with traditional drug delivery systems, stimulus-responsive nanosystems offer higher sensitivity, greater versatility, wider applicability, and enhanced biosafety. Recent research has made significant contributions towards designing stimulus-responsive polymer nanosystems for CVDs diagnosis and treatment. This review summarizes recent advances in this field by classifying stimulus-responsive polymer nanocarriers according to different responsiveness types and describing numerous stimuli relevant to these materials. Additionally, we discuss various applications of stimulus-responsive polymer nanomaterials in CVDs theranostics. We hope that this review will provide valuable insights into optimizing the design of stimulus-response polymers for accelerating their clinical application in diagnosing and treating CVDs.


Subject(s)
Cardiovascular Diseases , Theranostic Nanomedicine , Humans , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/therapy , Cardiovascular Diseases/drug therapy , Animals , Polymers/chemistry , Stimuli Responsive Polymers/chemistry , Reactive Oxygen Species/metabolism , Drug Carriers/chemistry , Nanostructures/chemistry , Nanoparticles/chemistry , Hydrogen-Ion Concentration
16.
Int J Nanomedicine ; 19: 7399-7414, 2024.
Article in English | MEDLINE | ID: mdl-39071500

ABSTRACT

Background: Influenza A (H1N1) virus is a highly contagious respiratory disease that causes severe illness and death. Vaccines and antiviral drugs are limited by viral variation and drug resistance, so developing efficient integrated theranostic options appears significant in anti-influenza virus infection. Methods: In this study, we designed and fabricated covalent organic framework (COF) based theranostic platforms (T705@DATA-COF-Pro), which was composed of an RNA polymerase inhibitor (favipiravir, T705), the carboxyl-enriched COF (DATA-COF) nano-carrier and Cy3-labeled single DNA (ssDNA) probe. Results: The multi-porosity COF core provided an excellent micro-environment and smooth delivery for T705. The ssDNA probe coating bound to the nucleic acids of H1N1 selectively, thus controlling drug release and allowing fluorescence imaging. The combination of COF and probe triggered the synergism, promoting drug further therapeutic outcomes. With the aid of T705@DATA-COF-Pro platforms, the H1N1-infected mouse models lightly achieved diagnosis and significantly prolonged survival. Conclusion: This research underscores the distinctive benefits and immense potential of COF materials in nano-preparations for virus infection, offering novel avenues for the detection and treatment of H1N1 virus infection.


Subject(s)
Antiviral Agents , Influenza A Virus, H1N1 Subtype , Metal-Organic Frameworks , Orthomyxoviridae Infections , Theranostic Nanomedicine , Influenza A Virus, H1N1 Subtype/drug effects , Animals , Theranostic Nanomedicine/methods , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Mice , Orthomyxoviridae Infections/drug therapy , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Humans , Amides/chemistry , Amides/pharmacology , Pyrazines/chemistry , Pyrazines/pharmacology , Pyrazines/pharmacokinetics , Pyrazines/therapeutic use , Influenza, Human/drug therapy , Mice, Inbred BALB C , Madin Darby Canine Kidney Cells , Drug Carriers/chemistry , Dogs , Drug Liberation
17.
Int J Pharm ; 661: 124456, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38986962

ABSTRACT

Indocyanine green is an FDA-approved fluorescent imaging dye used for determining cardiac output, hepatic function, liver blood flow, and retinal perfusion. It has been investigated preclinically in photoacoustic imaging and photothermal therapy (PTT); however, ICG photodegradation limits its biomedical applications. An aggregated form of ICG, known as J-aggregate (IJA), exhibits superior photoacoustic signals and thermal stability than the monomeric ICG. Nevertheless, IJA still suffers from low stability in the biological milieu, and short in vivo blood circulation. To address these limitations, a range of nanocarriers have been developed to enhance IJA stability and performance. This review focuses on IJA potentials and limitations, besides the recent development of IJA-loaded nanocarriers, particularly for cancer imaging and therapy.


Subject(s)
Indocyanine Green , Theranostic Nanomedicine , Indocyanine Green/administration & dosage , Humans , Animals , Theranostic Nanomedicine/methods , Neoplasms/therapy , Neoplasms/drug therapy , Neoplasms/diagnostic imaging , Nanoparticles , Fluorescent Dyes/chemistry , Fluorescent Dyes/administration & dosage , Photoacoustic Techniques/methods , Drug Carriers/chemistry
18.
Int J Pharm ; 661: 124401, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38986966

ABSTRACT

The technique of engineering drug delivery vehicles continues to develop, which bring enhancements in working more efficiently and minimizing side effects to make it more effective and safer. The intense capability of therapeutic agents to remain undamaged in a harsh extracellular environment is helpful to the success of drug development efforts. With this in mind, alterations of biopharmaceuticals with enhanced stability and decreased immunogenicity have been an increasingly active focus of such efforts. Red blood cells (RBCs), also known as erythrocytes have undergone extensive scrutiny as potential vehicles for drug delivery due to their remarkable attributes over the years of research. These include intrinsic biocompatibility, minimal immunogenicity, flexibility, and prolonged systemic circulation. Throughout the course of investigation, a diverse array of drug delivery platforms based on RBCs has emerged. These encompass genetically engineered RBCs, non-genetically modified RBCs, and RBC membrane-coated nanoparticles, each devised to cater to a range of biomedical objectives. Given their prevalence in the circulatory system, RBCs have gained significant attention for their potential to serve as biomimetic coatings for artificial nanocarriers. By virtue of their surface emulation capabilities and customizable core materials, nanocarriers mimicking these RBCs, hold considerable promise across a spectrum of applications, spanning drug delivery, imaging, phototherapy, immunomodulation, sensing, and detection. These multifaceted functionalities underscore the considerable therapeutic and diagnostic potential across various diseases. Our proposed review provides the synthesis of recent strides in the theranostic utilization of erythrocytes in the context of cancer. It also delves into the principal challenges and prospects intrinsic to this realm of research. The focal point of this review pertains to accentuating the significance of erythrocyte-based theranostic systems in combating cancer. Furthermore, it precisely records the latest and the most specific methodologies for tailoring the attributes of these biomimetic nanoscale formulations, attenuating various discoveries for the treatment and management of cancer.


Subject(s)
Biomimetics , Erythrocytes , Neoplasms , Theranostic Nanomedicine , Humans , Erythrocytes/drug effects , Neoplasms/therapy , Neoplasms/drug therapy , Theranostic Nanomedicine/methods , Animals , Biomimetics/methods , Drug Delivery Systems/methods , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Biomimetic Materials/chemistry , Nanoparticles , Drug Carriers/chemistry
19.
ACS Appl Mater Interfaces ; 16(30): 39005-39020, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39034639

ABSTRACT

Advanced photosensitizers for high-performance fluorescence imaging-guided photothermal therapy demand excellent near-infrared (NIR) brightness [molar absorption coefficient (ε) × quantum yield (QY)] and exceptional photothermal performance [ε × photothermal conversion efficiency (PCE)]. However, integrating high brightness and potent photothermal performance within a single molecule faces a formidable challenge. This article proposes a method to address this issue by preparing J-aggregate nanoparticles (NPs) using molecules with high ε. J-aggregates effectively improve QY and induce molecular emission redshift, while high ε molecules play a crucial role in improving the brightness and photothermal performance. By optimizing the molecular structure based on the pyrrolopyrrole cyanine (PPCy), precise control over the QY and PCE of PPCy J-aggregates is achieved. Ultimately, PDDO NPs exhibiting superior brightness (ε × QY = 3.32 × 104 M-1 cm-1) and photothermal performance (ε × PCE = 1.21 × 105 M-1 cm-1) are identified as high-performance photosensitizers. Notably, each parameter represents one of the highest levels among the reported fluorescence or photothermal probes to date. The in vivo studies demonstrate that PDDO NPs possess exceptional NIR imaging capabilities and remarkable photothermal tumor inhibition rates. This study provides innovative insights into the development of high-performance multifunctional photosensitizers.


Subject(s)
Nanoparticles , Photosensitizing Agents , Pyrroles , Theranostic Nanomedicine , Animals , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Mice , Pyrroles/chemistry , Pyrroles/pharmacology , Humans , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Infrared Rays , Photothermal Therapy , Carbocyanines/chemistry , Female , Mice, Inbred BALB C , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacology , Optical Imaging , Cell Line, Tumor , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Phototherapy
20.
ACS Appl Mater Interfaces ; 16(28): 35925-35935, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38950334

ABSTRACT

The development of efficient theranostic nanoagents for the precise diagnosis and targeted therapy of glioblastoma (GBM) remains a big challenge. Herein, we designed and developed porphyrin-based organic nanoparticles (PNP NPs) with strong emission in the near-infrared IIa window (NIR-IIa) for orthotopic GBM theranostics. PNP NPs possess favorable photoacoustic and photothermal properties, high photostability, and low toxicity. After modification with the RGD peptide, the obtained PNPD NPs exhibited enhanced blood-brain barrier (BBB) penetration capability and GBM targeting ability. NIR-IIa imaging was employed to monitor the in vivo biodistribution and accumulation of the nanoparticles, revealing a significant enhancement in penetration depth and signal-to-noise ratio. Both in vitro and in vivo results demonstrated that PNPD NPs effectively inhibited the proliferation of tumor cells and induced negligible side effects in normal brain tissues. In general, the work presented a kind of brain-targeted porphyrin-based NPs with NIR-IIa fluorescence for orthotopic glioblastoma theranostics, showing promising prospects for clinical translation.


Subject(s)
Glioblastoma , Nanoparticles , Porphyrins , Theranostic Nanomedicine , Glioblastoma/diagnostic imaging , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/metabolism , Animals , Nanoparticles/chemistry , Humans , Porphyrins/chemistry , Porphyrins/pharmacology , Mice , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Infrared Rays , Tissue Distribution , Blood-Brain Barrier/metabolism , Mice, Nude , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Mice, Inbred BALB C , Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL