Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters

Publication year range
1.
Nanotechnology ; 29(6): 065501, 2018 Feb 09.
Article in English | MEDLINE | ID: mdl-29226844

ABSTRACT

The logic device demultiplexer can convey a single input signal into one of multiple output channels. The choice of the output channel is controlled by a selector. Several molecules and biomolecules have been used to mimic the function of a demultiplexer. However, the practical application of logic devices still remains a big challenge. Herein, we design and construct an intelligent 1:2 demultiplexer as a theranostic device based on azobenzene (azo)-modified and DNA/Ag cluster-gated nanovehicles. The configuration of azo and the conformation of the DNA ensemble can be regulated by light irradiation and pH, respectively. The demultiplexer which uses light as the input and acid as the selector can emit red fluorescence or a release drug under different conditions. Depending on different cells, the intelligent logic device can select the mode of cellular imaging in healthy cells or tumor therapy in tumor cells. The study incorporates the logic gate with the theranostic device, paving the way for tangible applications of logic gates in the future.


Subject(s)
DNA/chemistry , Intracellular Space/chemistry , Nanoparticles/chemistry , Silver/chemistry , Theranostic Nanomedicine/instrumentation , Animals , Cell Death , Cell Survival , Endocytosis , HeLa Cells , Humans , Hydrogen-Ion Concentration , Light , Mice , NIH 3T3 Cells , Nanocomposites/chemistry , Nanocomposites/ultrastructure
2.
Biol Pharm Bull ; 41(4): 487-503, 2018 Apr 01.
Article in English | MEDLINE | ID: mdl-29332929

ABSTRACT

Detection of anomalous cells such as cancer cells from normal blood cells has the potential to contribute greatly to cancer diagnosis and therapy. Conventional methods for the detection of cancer cells are usually tedious and cumbersome. Herein, we report on the use of a particle size analyzer for the convenient size-based differentiation of cancer cells from normal cells. Measurements made using a particle size analyzer revealed that size parameters for cancer cells are significantly greater (e.g., inner diameter and width) than the corresponding values for normal cells (white blood cells (WBC), lymphocytes and splenocytes), with no significant difference in shape parameters (e.g., circularity and convexity). The inner diameter of many cancer cell lines is greater than 10 µm, in contrast to normal cells. For the detection of WBC having similar size to that of cancer cells, we developed a PC software "Cancer Cell Finder" that differentiates them from cancer cells based on brightness stationary points on a cell surface. Furthermore, the aforementioned method was validated for cancer cell/clusters detection in spiked mouse blood samples (a B16 melanoma mouse xenograft model) and circulating tumor cell cluster-like particles in the cat and dog (diagnosed with cancer) blood samples. These results provide insights into the possible applicability of the use of a particle size analyzer in conjunction with PC software for the convenient detection of cancer cells in experimental and clinical samples for theranostics.


Subject(s)
Breast Neoplasms/pathology , Colorectal Neoplasms/pathology , Lung Neoplasms/pathology , Melanoma, Experimental/pathology , Prostatic Neoplasms/pathology , Animals , Breast Neoplasms/blood , Breast Neoplasms/diagnosis , Cats , Cell Line, Tumor , Cell Shape , Cell Size , Colorectal Neoplasms/blood , Colorectal Neoplasms/diagnosis , Dogs , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Leukocytes/cytology , Leukocytes/pathology , Lung Neoplasms/blood , Lung Neoplasms/diagnosis , Lymphocytes/cytology , Lymphocytes/pathology , Male , Melanoma, Experimental/blood , Melanoma, Experimental/diagnosis , Mice, Inbred C57BL , Prostatic Neoplasms/blood , Prostatic Neoplasms/diagnosis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Software , Spleen/cytology , Spleen/pathology , Surface Properties , Theranostic Nanomedicine/instrumentation , Theranostic Nanomedicine/methods
3.
J Nanobiotechnology ; 16(1): 30, 2018 Mar 27.
Article in English | MEDLINE | ID: mdl-29587764

ABSTRACT

BACKGROUND: The rational design of theranostic nanoprobe to present responsive effect of therapeutic potency and enhanced diagnostic imaging in tumor milieu plays a vital role for efficient personalized cancer therapy and other biomedical applications. We aimed to afford a potential strategy to pose both T1- and T2-weighted MRI functions, and thereby realizing imaging guided drug delivery and targeted therapy. RESULTS: Theranostic nanocomposites Mn-porphyrin&Fe3O4@SiO2@PAA-cRGD were fabricated and characterized, and the nanocomposites were effectively used in T1- and T2-weighted MRI and pH-responsive drug release. Fluorescent imaging also showed that the nanocomposites specifically accumulated in lung cancer cells by a receptor-mediated process, and were nontoxic to normal cells. The r2/r1 ratio was 20.6 in neutral pH 7.4, which decreased to 7.7 in acidic pH 5.0, suggesting the NCs could act as an ideal T1/T2 dual-mode contrast agent at acidic environments of tumor. For in vivo MRI, T1 and T2 relaxation was significantly accelerated to 55 and 37%, respectively, in the tumor after i.v. injection of nanocomposites. CONCLUSION: The synthesized nanocomposites exhibited highly sensitive MRI contrast function no matter in solution, cells or in vivo by synergistically enhancing positive and negative magnetic resonance imaging signals. The nanocomposites showed great potential for integrating imaging diagnosis and drug controlled release into one composition and providing real-time imaging with greatly enhanced diagnostic accuracy during targeted therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Contrast Media/chemistry , Doxorubicin/pharmacokinetics , Lung Neoplasms/diagnostic imaging , Magnetic Resonance Imaging/methods , Nanocomposites/chemistry , Theranostic Nanomedicine/methods , A549 Cells , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Contrast Media/pharmacokinetics , Doxorubicin/pharmacology , Drug Compounding/methods , Ferrosoferric Oxide/chemistry , Humans , Hydrogen-Ion Concentration , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Male , Manganese/chemistry , Mice , Mice, Nude , Nanocomposites/administration & dosage , Nanocomposites/ultrastructure , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Oligopeptides/chemistry , Porphyrins/chemistry , Silicon Dioxide/chemistry , Theranostic Nanomedicine/instrumentation , Xenograft Model Antitumor Assays
4.
Biochim Biophys Acta Gen Subj ; 1861(6): 1530-1544, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28130158

ABSTRACT

BACKGROUND: Theranostics, a fusion of two key parts of modern medicine - diagnostics and therapy of the organism's disorders, promises to bring the efficacy of medical treatment to a fundamentally new level and to become the basis of personalized medicine. Extrapolating today's progress in the field of smart materials to the long-run prospect, we can imagine future intelligent agents capable of performing complex analysis of different physiological factors inside the living organism and implementing a built-in program thereby triggering a series of therapeutic actions. These agents, by analogy with their macroscopic counterparts, can be called nanorobots. It is quite obscure what these devices are going to look like but they will be more or less based on today's achievements in nanobiotechnology. SCOPE OF REVIEW: The present Review is an attempt to systematize highly diverse nanomaterials, which may potentially serve as modules for theranostic nanorobotics, e.g., nanomotors, sensing units, and payload carriers. MAJOR CONCLUSIONS: Biocomputing-based sensing, externally actuated or chemically "fueled" autonomous movement, swarm inter-agent communication behavior are just a few inspiring examples that nanobiotechnology can offer today for construction of truly intelligent drug delivery systems. GENERAL SIGNIFICANCE: The progress of smart nanomaterials toward fully autonomous drug delivery nanorobots is an exciting prospect for disease treatment. Synergistic combination of the available approaches and their further development may produce intelligent drugs of unmatched functionality.


Subject(s)
Biosensing Techniques/instrumentation , Drug Carriers/chemistry , Drug Delivery Systems/instrumentation , Nanostructures/chemistry , Pharmaceutical Preparations/chemistry , Theranostic Nanomedicine/instrumentation , Animals , Drug Carriers/classification , Drug Compounding , Drug Delivery Systems/methods , Humans , Nanostructures/classification , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/classification , Terminology as Topic , Theranostic Nanomedicine/methods
5.
Drug Resist Updat ; 27: 39-58, 2016 07.
Article in English | MEDLINE | ID: mdl-27449597

ABSTRACT

The progress in medical research has led to the understanding that cancer is a large group of heterogeneous diseases, with high variability between and within individuals. This variability sprouted the ambitious goal to improve therapeutic outcomes, while minimizing drug adverse effects through stratification of patients by the differences in their disease markers, in a personalized manner, as opposed to the strategy of "one therapy fits all". Nanotheranostics, composed of nanoparticles (NPs) carrying therapeutic and/or diagnostics probes, have the potential to revolutionize personalized medicine. There are different modalities to combine these two distinct fields into one system for a synergistic outcome. The addition of a nanocarrier to a theranostic system holds great promise. Nanocarriers possess high surface area, enabling sophisticated functionalization with imaging agents, thus gaining enhanced diagnostic ability in real-time. Yet, most of the FDA-approved theranostic approaches are based on small molecules. The theranostic approaches that are reviewed herein are paving the road towards personalized medicine through all stages of patient care: starting from screening and diagnostics, proceeding to treatment and ending with treatment follow-up. Our current review provides a broad background and highlights new insights for the rational design of theranostic nanosystems for desired therapeutic niches, while summoning the hurdles on their way to become first-line diagnostics and therapeutics for cancer patients.


Subject(s)
Diagnostic Imaging/methods , Drug Delivery Systems/methods , Neoplasms/diagnostic imaging , Precision Medicine/methods , Theranostic Nanomedicine/methods , Clinical Trials as Topic , Diagnostic Imaging/instrumentation , Drug Monitoring/instrumentation , Drug Monitoring/methods , Humans , Microbubbles/therapeutic use , Nanoparticles/administration & dosage , Neoplasm Staging , Neoplasms/pathology , Neoplasms/therapy , Photochemotherapy/methods , Precision Medicine/instrumentation , Radiation-Sensitizing Agents/therapeutic use , Radiotherapy, Image-Guided/methods , Theranostic Nanomedicine/instrumentation
6.
Vopr Onkol ; 62(5): 559-569, 2016.
Article in Russian | MEDLINE | ID: mdl-30695578

ABSTRACT

The review discusses the relevance of creating original theranostic apparatus for photodynamic therapy (PDT) and fluorescent diagnostics (FD) to fulfill the needs and meet the objectives of oncology field specialists. Increased accuracy of diagnosis and improvement of treatment outcomes is possible only in case of the right combination of photosensitizer (PS) with the light source with a wavelength at which the maximum absorption of the PS is observed. The absence of medical devices combining functions of PDT and FD makes the development of the domestic theranostic apparatus for oncology an extremely important task.


Subject(s)
Neoplasms/diagnosis , Neoplasms/drug therapy , Optical Imaging/instrumentation , Photochemotherapy/instrumentation , Theranostic Nanomedicine/instrumentation , Humans , Optical Imaging/methods , Photochemotherapy/methods , Theranostic Nanomedicine/methods
8.
Int J Nanomedicine ; 19: 5479-5492, 2024.
Article in English | MEDLINE | ID: mdl-38863646

ABSTRACT

Background: In recent years, PD-L1 has been primarily utilized as an immune checkpoint marker in cancer immunotherapy. However, due to tumor heterogeneity, the response rate to such therapies often falls short of expectations. In addition to its role in immunotherapy, PD-L1 serves as a specific target on the surface of tumor cells for targeted diagnostic and therapeutic interventions. There is an absence of a fully developed PD-L1-targeted diagnostic and therapeutic probe for clinical use, which constrains the exploration and clinical exploitation of this target. Methods and Results: In this study, we engineered a PD-L1-targeted probe with multimodal imaging and dual therapeutic functionalities utilizing organic melanin nanoparticles. Functionalization with the WL12-SH peptide endowed the nanoprobe with specific targeting capabilities. Subsequent radiolabeling with 89Zr (half-life: 100.8 hours) and chelation of Mn2+ ions afforded the probe the capacity for simultaneous PET and MRI imaging modalities. Cellular uptake assays revealed pronounced specificity, with -positive cells exhibiting significantly higher uptake than -negative counterparts (p < 0.05). Dual-modal PET/MRI imaging delineated rapid and sustained accumulation at the neoplastic site, yielding tumor-to-non-tumor (T/NT) signal ratios at 24 hours post-injection of 16.67±3.45 for PET and 6.63±0.64 for MRI, respectively. We conjugated the therapeutic radionuclide 131I (half-life: 8.02 days) to the construct and combined low-dose radiotherapy and photothermal treatment (PTT), culminating in superior antitumor efficacy while preserving a high safety profile. The tumors in the cohort receiving the dual-modality therapy exhibited significantly reduced volume and weight compared to those in the control and monotherapy groups. Conclusion: We developed and applied a novel -targeted multimodal theranostic nanoprobe, characterized by its high specificity and superior imaging capabilities as demonstrated in PET/MRI modalities. Furthermore, this nanoprobe facilitates potent therapeutic efficacy at lower radionuclide doses when used in conjunction with PTT.


Subject(s)
B7-H1 Antigen , Immunotherapy , Multimodal Imaging , Theranostic Nanomedicine , Animals , Female , Humans , Mice , B7-H1 Antigen/metabolism , Cell Line, Tumor , Immunotherapy/methods , Magnetic Resonance Imaging/methods , Melanins/chemistry , Multimodal Imaging/instrumentation , Multimodal Imaging/methods , Nanoparticles/chemistry , Positron-Emission Tomography/methods , Radioisotopes/chemistry , Theranostic Nanomedicine/instrumentation , Theranostic Nanomedicine/methods , Zirconium/chemistry
10.
Small Methods ; 6(2): e2101217, 2022 02.
Article in English | MEDLINE | ID: mdl-34957704

ABSTRACT

Nanotheranostic materials (NTMs) shed light on the mechanisms responsible for complex diseases such as cancer because they enable making a diagnosis, monitoring the disease progression, and applying a targeted therapy simultaneously. However, several issues such as the reproducibility and mass production of NTMs hamper their application for clinical practice. To address these issues and facilitate the clinical application of NTMs, microfluidic systems have been increasingly used. This perspective provides a glimpse into the current state-of-art of NTM research, emphasizing the methods currently employed at each development stage of NTMs and the related open problems. This work reviews microfluidic technologies used to develop NTMs, ranging from the fabrication and testing of a single NTM up to their manufacturing on a large scale. Ultimately, a step-by-step vision on the future development of NTMs for clinical practice enabled by microfluidics techniques is provided.


Subject(s)
Lab-On-A-Chip Devices , Theranostic Nanomedicine/instrumentation , Animals , Humans , Theranostic Nanomedicine/methods
11.
Biomed Mater ; 16(2): 022009, 2021 02 24.
Article in English | MEDLINE | ID: mdl-33307545

ABSTRACT

Extracellular vesicles (EVs) are cell secretory native components with long-circulation, good biocompatibility, and physiologic barriers cross ability. EVs derived from different donor cells inherit varying characteristics and functions from their original cells and are favorable to serve as vectors for diagnosing and treating various diseases. However, EVs nanotheranostics are still in their infancy because of their limited accumulation at lesion sites and compromised therapy efficiency. Hence, engineering modification of EVs is usually needed to further enhance their stability, biological activity, and lesion-targeting capacity. Herein, we overview the characteristics of EVs from different sources, as well as the latest developments of surface engineering and cargo loading methods. We also focus especially on advances in EVs-based disease theranostics. At the end of the review, we predict the obstacles and prospects of the future clinical application of EVs.


Subject(s)
Cell Communication , Drug Delivery Systems , Nanomedicine/methods , Theranostic Nanomedicine/methods , Tissue Engineering/methods , Animals , Dendritic Cells/cytology , Extracellular Vesicles/metabolism , Humans , Killer Cells, Natural/metabolism , Macrophages/metabolism , Precision Medicine , Stem Cells/cytology , T-Lymphocytes/metabolism , Theranostic Nanomedicine/instrumentation , Tissue Engineering/instrumentation
12.
Nat Commun ; 12(1): 7149, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34887404

ABSTRACT

Radiotherapy is an important therapeutic strategy for cancer treatment through direct damage to cancer cells and augmentation of antitumor immune responses. However, the efficacy of radiotherapy is limited by hypoxia-mediated radioresistance and immunosuppression in tumor microenvironment. Here, we construct a stabilized theranostic nanoprobe based on quantum dots emitting in the near-infrared IIb (NIR-IIb, 1,500-1,700 nm) window modified by catalase, arginine-glycine-aspartate peptides and poly(ethylene glycol). We demonstrate that the nanoprobes effectively aggregate in the tumor site to locate the tumor region, thereby realizing precision radiotherapy with few side-effects. In addition, nanoprobes relieve intratumoral hypoxia and reduce the tumor infiltration of immunosuppressive cells. Moreover, the nanoprobes promote the immunogenic cell death of cancer cells to trigger the activation of dendritic cells and enhance T cell-mediated antitumor immunity to inhibit tumor metastasis. Collectively, the nanoprobe-mediated immunogenic radiotherapy can boost the abscopal effect to inhibit tumor metastasis and prolong survival.


Subject(s)
Nanoparticles/chemistry , Neoplasms/radiotherapy , Neoplasms/therapy , Theranostic Nanomedicine/methods , Animals , Cell Line, Tumor , Combined Modality Therapy , Dendritic Cells/immunology , Dendritic Cells/radiation effects , Female , Humans , Immunotherapy , Infrared Rays , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/mortality , Quantum Dots/chemistry , Radiotherapy/instrumentation , Radiotherapy/methods , T-Lymphocytes/immunology , T-Lymphocytes/radiation effects , Theranostic Nanomedicine/instrumentation , Tumor Microenvironment/radiation effects
13.
Macromol Biosci ; 21(6): e2000424, 2021 06.
Article in English | MEDLINE | ID: mdl-33811465

ABSTRACT

How to precisely detect and effectively cure cancer which is defined as precise nanomedicine has drawn great attention worldwide. Polymeric nanoreactors which can in situ catalyze inert species into activated ones, can greatly increase imaging quality and enhance therapeutic effects along with decreased background interference and reduced serious side effects. After a brief introduction, the design and preparation of polymeric nanoreactors are discussed from the following aspects, that is, solvent-switch, pH-tuning, film rehydration, hard template, electrostatic interaction, and polymerization-induced self-assembly (PISA). Subsequently, the biomedical applications of these nanoreactors in the fields of cancer imaging, cancer therapy, and cancer theranostics are highlighted. The last but not least, conclusions and future perspectives about polymeric nanoreactors are given. It is believed that polymeric nanoreactors can bring a great opportunity for future fabrication and clinical translation of precise nanomedicine.


Subject(s)
Drug Carriers , Nanostructures/chemistry , Neoplasms/therapy , Polymers/chemical synthesis , Precision Medicine/methods , Theranostic Nanomedicine/methods , A549 Cells , Animals , Bioreactors , Humans , Hydrogen-Ion Concentration , Membranes, Artificial , Mice , Nanostructures/administration & dosage , Nanostructures/ultrastructure , Neoplasms/metabolism , Neoplasms/pathology , Polymers/pharmacokinetics , Precision Medicine/instrumentation , Solvents/chemistry , Static Electricity , Surface-Active Agents/chemistry , Surface-Active Agents/pharmacokinetics , Theranostic Nanomedicine/instrumentation , Xenograft Model Antitumor Assays
14.
Theranostics ; 11(17): 8448-8463, 2021.
Article in English | MEDLINE | ID: mdl-34373752

ABSTRACT

Rationale: Near-Infrared persistent luminescence (NIR-PL) nanomaterials that can continually emit low-energy photons after ceasing excitation has emerged as a new generation of theranostic nanoparticle drug delivery systems (NDDSs) for imaging-guided cancer therapy, which stems from their special ability to completely avoid tissue autofluorescence interference. However, unresponsive diagnostic capability, inefficient drug delivery, and poor biodegradability limit the efficacy of most reported NIR-PL-based NDDSs. Methods: Herein, a multifaceted tumor microenvironment (TME)-degradable theranostic drug delivery nanocapsule based on an ultrasmall persistent phosphor with a hollow mesoporous manganese-doped, DOX-loaded silica shell (Mn-ZGOCS-PEG) is developed to overcome the above drawbacks. Results: We demonstrate that the well-designed nanocapsule enables tumor-responsive controlled drug release with ameliorated therapeutic efficacy, TME-responsive autofluorescence interference-free NIR-PL tracing, and manganese-enhanced magnetic resonance (Mn-MR) monitoring for practical dual-modality image-guided antitumor treatment in vivo. Conclusion: Our results indicate that Mn-ZGOCS-PEG nanocapsules enable tumor-targeting augmented chemotherapy under the guidance of TME-responsive dual-MR/NIR-PL-modality imaging in vivo. We believe that our work provides a new paradigm for the development of smart NIR-PL-based NDDSs with ultrasensitive multimodal diagnostic capability, enhanced anticancer effect, and efficient biodegradability.


Subject(s)
Manganese/chemistry , Theranostic Nanomedicine/instrumentation , Theranostic Nanomedicine/methods , Absorbable Implants , Animals , Cell Line, Tumor , Doxorubicin/therapeutic use , Drug Delivery Systems/methods , Drug Therapy/methods , Humans , Infrared Rays , Infusion Pumps, Implantable , Luminescence , Magnetic Resonance Imaging/methods , Male , Manganese/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Nanocapsules , Nanoparticles/therapeutic use , Photochemotherapy/methods , Spectroscopy, Near-Infrared/methods , Tumor Microenvironment/drug effects
15.
Methods Mol Biol ; 2312: 141-158, 2021.
Article in English | MEDLINE | ID: mdl-34228289

ABSTRACT

With the development of mobile communication technology, smartphones have been used in point-of-care technologies (POCTs) as an important part of telemedicine. Using a multidisciplinary design principle coupling electrical engineering, software development, synthetic biology, and optogenetics, the investigators developed a smartphone-controlled semiautomatic theranostic system that regulates blood glucose homeostasis in diabetic mice in an ultraremote-control manner. The present chapter describes how the investigators tailor-designed the implant architecture "HydrogeLED," which is capable of coharboring a designer-cell-carrying alginate hydrogel and wirelessly powered far-red light LEDs. Using diabetes mellitus as a model disease, the in vivo expression of insulin or human glucagon-like peptide 1 (shGLP-1) from HydrogeLED implants could be controlled not only by pre-set ECNU-TeleMed programs, but also by a custom-engineered Bluetooth-active glucometer in a semiautomatic and glycemia-dependent manner. As a result, blood glucose homeostasis was semiautomatically maintained in diabetic mice through the smartphone-controlled semiautomatic theranostic system. By combining digital signals with optogenetically engineered cells, the present study provides a new method for the integrated diagnosis and treatment of diseases.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Experimental/therapy , Glycemic Control/instrumentation , Optogenetics/instrumentation , Smartphone , Telemedicine/instrumentation , Theranostic Nanomedicine/instrumentation , Wireless Technology/instrumentation , Alginates/chemistry , Animals , Biomarkers/blood , Blood Glucose Self-Monitoring/instrumentation , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/diagnosis , Diabetes Mellitus, Experimental/genetics , Gene Expression Regulation/radiation effects , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , HEK293 Cells , Homeostasis , Humans , Hydrogels , Insulin/genetics , Insulin/metabolism , Light , Male , Mice, Inbred C57BL , Mobile Applications
16.
Acta Biomater ; 101: 372-383, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31622780

ABSTRACT

Cytokines mediate and control immune and inflammatory responses. Complex interactions exist among cytokines, inflammation, and the innate and adaptive immune responses in maintaining homeostasis, health, and well-being. On-demand, local delivery of anti-inflammatory drugs to target tissues provides an approach for more effective drug dosing while reducing the adverse effects of systemic drug delivery. This work demonstrates a proof-of-concept theranostic approach for inflammation based on analyte-kissing induced signaling, whereby a drug (in this report, aspirin) can be released upon the detection of a target level of a proinflammatory cytokine (i.e., interferon-γ (IFN-γ)) in real time. The structure-switching aptamer-based biosensor described here is capable of quantitatively and dynamically detecting IFN-γ both in vitro and in vivo with a sensitivity of 10 pg mL-1. Moreover, the released aspirin triggered by the immunoregulatory cytokine IFN-γ is able to inhibit inflammation in a rat model, and the release of aspirin can be quantitatively controlled. The data reported here provide a new and promising strategy for the in vivo detection of proinflammatory cytokines and the subsequent therapeutic delivery of anti-inflammatory molecules. This universal theranostic platform is expected to have great potential for patient-specific personalized medicine. STATEMENT OF SIGNIFICANCE: We developed an adaptive in vivo sensing device whereby a drug, aspirin, can be released upon the detection of a proinflammatory cytokine, interferon-γ (IFN-γ), in real time with a sensitivity of 10 pg mL-1. Moreover, the aspirin triggered by IFN-γ depressed inflammation in the rat model and was delivered indirectly through blood and cerebrospinal fluid or directly to the inflammation tissue or organ without adverse gastrointestinal effects observed in the liver and kidney. We envision that, for the first time, patients with chronic inflammatory disease can receive the right intervention and treatment at the right time. Additionally, this technology may empower patients to monitor their personalized health and disease management program, allowing real-time diagnostics, disease monitoring, and precise and effective treatments.


Subject(s)
Aspirin/pharmacology , Inflammation/pathology , Interferon-gamma/pharmacology , Theranostic Nanomedicine/instrumentation , Animals , Aptamers, Peptide/chemistry , Biosensing Techniques , Carbon/chemistry , Cellular Microenvironment/drug effects , Electrochemical Techniques , Glass/chemistry , Humans , Lung/pathology , Male , Photoelectron Spectroscopy , Rats, Sprague-Dawley , Streptavidin/chemistry
17.
IEEE Trans Nanobioscience ; 19(4): 627-632, 2020 10.
Article in English | MEDLINE | ID: mdl-32746332

ABSTRACT

Ultrashort peptides (USPs), composed of three to seven amino acids, can self-assemble into nanofibers in pure water. Here, using hydrodynamic focusing and a solvent exchange method on a microfluidic setup, we convert these nanofibers into globular nanoparticles with excellent dimensional control and polydispersity. Thanks to USP nanocarriers' structure, different drugs can be loaded. We used Curcumin as a model drug to evaluate the performance of USP nanocarriers as a novel drug delivery vehicle. These nanoparticles can efficiently cross the cell membrane and possess nonlinear optical properties. Therefore, we envisage USP nanoparticles as promising future theranostic nanocarriers.


Subject(s)
Drug Carriers , Microfluidic Analytical Techniques/instrumentation , Nanoparticles/chemistry , Peptides , Theranostic Nanomedicine , Curcumin/chemistry , Curcumin/pharmacokinetics , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Equipment Design , HeLa Cells , Humans , Peptides/chemistry , Peptides/pharmacokinetics , Solvents/chemistry , Theranostic Nanomedicine/instrumentation , Theranostic Nanomedicine/methods
18.
J Nucl Med ; 61(1): 80-88, 2020 01.
Article in English | MEDLINE | ID: mdl-31253744

ABSTRACT

Targeted radiopharmaceutical therapy (TRT) using α-particle radiation is a promising approach for treating both large and micrometastatic lesions. We developed prostate-specific membrane antigen (PSMA)-targeted low-molecular-weight agents for 212Pb-based TRT of patients with prostate cancer (PC) by evaluating the matching γ-emitting surrogate, 203Pb. Methods: Five rationally designed low-molecular-weight ligands (L1-L5) were synthesized using the lysine-urea-glutamate scaffold, and PSMA inhibition constants were determined. Tissue biodistribution and SPECT/CT imaging of 203Pb-L1-203Pb-L5 were performed on mice bearing PSMA(+) PC3 PIP and PSMA(-) PC3 flu flank xenografts. The absorbed radiation dose of the corresponding 212Pb-labeled analogs was determined using the biodistribution data. Antitumor efficacy of 212Pb-L2 was evaluated in PSMA(+) PC3 PIP and PSMA(-) PC3 flu tumor models and in the PSMA(+) luciferase-expressing micrometastatic model. 212Pb-L2 was also evaluated for dose-escalated, long-term toxicity. Results: All new ligands were obtained in high yield and purity. PSMA inhibitory activities ranged from 0.10 to 17 nM. 203Pb-L1-203Pb-L5 were synthesized in high radiochemical yield and specific activity. Whole-body clearance of 203Pb-L1-203Pb-L5 was fast. The absorbed dose coefficients (mGy/kBq) of the tumor and kidneys were highest for 203Pb-L5 (31.0, 15.2) and lowest for 203Pb-L2 (8.0, 4.2). The tumor-to-kidney absorbed dose ratio was higher for 203Pb-L3 (3.2) and 203Pb-L4 (3.6) than for the other agents, but with lower tumor-to-blood ratios. PSMA(+) tumor lesions were visualized through SPECT/CT as early as 0.5 h after injection. A proof-of-concept therapy study with a single administration of 212Pb-L2 demonstrated dose-dependent inhibition of tumor growth in the PSMA(+) flank tumor model. 212Pb-L2 also demonstrated an increased survival benefit in the micrometastatic model compared with 177Lu-PSMA-617. Long-term toxicity studies in healthy, immunocompetent CD-1 mice revealed kidney as the dose-limiting organ. Conclusion:203Pb-L1-203Pb-L5 demonstrated favorable pharmacokinetics for 212Pb-based TRT. The antitumor efficacy of 212Pb-L2 supports the corresponding 203Pb/212Pb theranostic pair for PSMA-based α-particle TRT in advanced PC.


Subject(s)
Lead Radioisotopes/pharmacokinetics , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/radiotherapy , Radiopharmaceuticals/pharmacokinetics , Theranostic Nanomedicine/instrumentation , Alpha Particles , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Humans , Kaplan-Meier Estimate , Kidney/diagnostic imaging , Ligands , Male , Maximum Tolerated Dose , Mice , Neoplasm Metastasis , Proteasome Endopeptidase Complex/analysis , Radiation Dosage , Radiometry , Single Photon Emission Computed Tomography Computed Tomography , Theranostic Nanomedicine/methods , Tumor Protein, Translationally-Controlled 1
19.
ACS Appl Mater Interfaces ; 12(3): 3445-3452, 2020 Jan 22.
Article in English | MEDLINE | ID: mdl-31922399

ABSTRACT

For effective treatment of ischemic cerebral thrombosis, it is of great significance to find a facile way in assessing the early damage of blood-brain barrier (BBB) after ischemic stroke during thrombolysis by integrating thrombolytic agents with fluorescent materials. Herein, a novel type of protein-carbon dot  nanohybrids is reported by the incorporation of carbon dots on thrombolytic agents through covalent linkage. Both in vitro and ex vivo fluorescence imaging measurements have demonstrated remarkable imaging effects in the brain of transient middle cerebral artery occlusion mice. Besides, the outstanding thrombolytic capacity of the nanohybrids was determined by in vitro thrombolysis tests. As one of the few reports of the construction of thrombolytic agents and fluorescent nanomaterials, the nanohybrids retain thrombolysis ability and fluorescent traceability simultaneously. It may provide a promising indicator for early BBB damage and thrombolytic agent distribution to estimate the possibility of symptomatic intracranial hemorrhage after thrombolysis and supply tissue window evidence for clinical thrombolytic agent application.


Subject(s)
Blood-Brain Barrier/drug effects , Carbon/chemistry , Fibrinolytic Agents/administration & dosage , Nanostructures/chemistry , Stroke/drug therapy , Theranostic Nanomedicine/methods , Urokinase-Type Plasminogen Activator/administration & dosage , Animals , Fibrinolytic Agents/chemistry , Humans , Male , Mice , Mice, Inbred BALB C , Stroke/diagnostic imaging , Theranostic Nanomedicine/instrumentation , Urokinase-Type Plasminogen Activator/chemistry
20.
Curr Drug Discov Technol ; 17(5): 661-669, 2020.
Article in English | MEDLINE | ID: mdl-30973109

ABSTRACT

BACKGROUND: Detailed photochemical and photocytotoxicity studies of two new porphyrins: 5,10,15,20-meso-tetrakis-(4-acetoxy-3-methoxyphenyl) porphyrin (P2.1) and 5-(4-hydroxy-3- methoxyphenyl)-10,15,20-tris-(4-acetoxy-3-methoxyphenyl)porphyrin (P2.2) are reported, as potential candidates for theranostics. For powdered samples of P2.1 and P2.2 adsorbed onto a powdered biocompatible substrate, polyethylene glycol (PEG), a concentration study was performed, correlating the fluorescence emission intensity with sample absorption to determine the useful concentration range for photodynamic therapy of cancer (PDT) in which aggregation does not occur. Cytotoxicity studies were performed in dark and illuminated conditions. METHODS: The laser induced luminescence set-up is home-made, a N2 laser is used as the excitation source and a time gated charged-coupled device (ICCD) as the detector. Fluorescence lifetime determinations were made using pulsed light sources from the excitation LEDs and measures of the fluorescence intensities at different time delays after the excitation pulse. The singlet oxygen formation quantum yields ΦΔ measurements were obtained by comparing the total area of the emission spectra for the reference compound and also for the samples under study in the same solvent and with the same optical density at the excitation wavelength (405 nm). An integrating sphere for relative and absolute measurements was used in this work as an alternative methodology to obtain the values for the fluorescence emission quantum yields (ΦF) of the adsorbed porphyrin under study. The cytotoxicity evaluation was made in the dark and under irradiation, using four different human tumor cell lines and one non-tumor primary cell culture. RESULTS: In order to establish the useful range of concentrations of the sensitizer for PDT, and due to the use of powdered samples, a special methodology was needed: the variations of the fluorescence lifetimes and fluorescence quantum yields were evaluated as a function of the concentration of the dye, measured by (1-R)*fdye. Both ΦF and τF are constant in the range from 0.002 to about 0.050 µmol g-1, and only after that a concentration quenching effect becomes visible, decreasing both ΦF and τF. This methodology is based in the correlations established between the Remission Function values and ΦF and τF obtained for increasing values of the sensitizer concentrations. CONCLUSIONS: The study of the aggregation effects of P2.1 and P2.2 porphyrins into a PEG matrix allowed us to determine the usable concentration range for photodynamic therapy use, where the aggregation of porphyrins decreases, therefore reducing the PDT action. The use of an integrating sphere for relative and absolute measurements of fluorescence quantum yields and also the lifetime studies as a function of the dye loading confirms the useful range for the use of P2.1 and P2.2 in PEG as powdered samples. The determination of the GI50, the porphyrin concentration which inhibits 50% of the cell growth, evidences that P2.2, the A3B porphyrin overtakes P2.1 (the A4 porphyrin) in terms of PDT efficiency and both porphyrins are much better PDT agents than the unsubstituted porphyrin, TPP. These data clearly show that porphyrins P2.2 and P2.1 exhibit an excellent behaviour in terms of its photocytotoxicity. These results encourage us to pursuit in the study of this family of porphyrins in which a balance of hydrophobic versus hydrophilic substituents in the phenyl group was achieved.


Subject(s)
Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Porphyrins/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Carriers/chemistry , Drug Evaluation, Preclinical , Humans , Lasers, Gas , Nanoparticles/chemistry , Neoplasms/pathology , Photochemotherapy/instrumentation , Photosensitizing Agents/pharmacokinetics , Polyethylene Glycols/chemistry , Porphyrins/pharmacokinetics , Theranostic Nanomedicine/instrumentation , Theranostic Nanomedicine/methods , Tissue Distribution/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL