Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 11.153
Filter
1.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 46(4): 610-618, 2024 Aug.
Article in Chinese | MEDLINE | ID: mdl-39223026

ABSTRACT

Immune checkpoint inhibitors have shown remarkable benefits in the treatment of solid tumors,while the occurrence of atypical response patterns and immune-related adverse events during treatment challenges the accuracy of therapeutic evaluation.Medical imaging is crucial for the evaluation of immunotherapy.It enables the assessment of treatment efficacy via both morphological and functional ways and offers unique a predictive value when being combined with artificial intelligence.Here we review the recent research progress in imaging-based evaluation of solid tumors treated with immune checkpoint inhibitors.


Subject(s)
Immune Checkpoint Inhibitors , Immunotherapy , Neoplasms , Humans , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Neoplasms/immunology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Artificial Intelligence
2.
Clin Transl Sci ; 17(9): e70001, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39206604

ABSTRACT

Traditional cancer classification based on organ of origin and histology is increasingly at odds with precision oncology. Tumors in different organs can share molecular features, while those in the same organ can be heterogeneous. This disconnect impacts clinical trials, drug development, and patient care. Recent advances in artificial intelligence (AI), particularly machine learning and deep learning, offer promising avenues for reclassifying cancers through comprehensive integration of molecular, histopathological, imaging, and clinical characteristics. AI-driven approaches have the potential to reveal novel cancer subtypes, identify new prognostic variables, and guide more precise treatment strategies for improving patient outcomes.


Subject(s)
Artificial Intelligence , Neoplasms , Humans , Neoplasms/classification , Neoplasms/pathology , Neoplasms/diagnosis , Neoplasms/diagnostic imaging , Precision Medicine/methods , Deep Learning , Algorithms , Machine Learning , Prognosis
3.
ACS Nano ; 18(35): 24295-24305, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39164203

ABSTRACT

Accurately distinguishing tumor cells from normal cells is a key issue in tumor diagnosis, evaluation, and treatment. Fluorescence-based immunohistochemistry as the standard method faces the inherent challenges of the heterogeneity of tumor cells and the lack of big data analysis of probing images. Here, we have demonstrated a machine learning-driven imaging method for rapid pathological diagnosis of five types of cancers (breast, colon, liver, lung, and stomach) using a perovskite nanocrystal probe. After conducting the bioanalysis of survivin expression in five different cancers, high-efficiency perovskite nanocrystal probes modified with the survivin antibody can recognize the cancer tissue section at the single cell level. The tumor to normal (T/N) ratio is 10.3-fold higher than that of a conventional fluorescent probe, which can successfully differentiate between tumors and adjacent normal tissues within 10 min. The features of the fluorescence intensity and pathological texture morphology have been extracted and analyzed from 1000 fluorescence images by machine learning. The final integrated decision model makes the area under the receiver operating characteristic curve (area under the curve) value of machine learning classification of breast, colon, liver, lung, and stomach above 90% while predicting the tumor organ of 92% of positive patients. This method demonstrates a high T/N ratio probe in the precise diagnosis of multiple cancers, which will be good for improving the accuracy of surgical resection and reducing cancer mortality.


Subject(s)
Calcium Compounds , Machine Learning , Neoplasms , Oxides , Titanium , Humans , Titanium/chemistry , Calcium Compounds/chemistry , Neoplasms/diagnosis , Neoplasms/pathology , Neoplasms/diagnostic imaging , Oxides/chemistry , Nanoparticles/chemistry , Optical Imaging , Fluorescent Dyes/chemistry
4.
Technol Cancer Res Treat ; 23: 15330338241273149, 2024.
Article in English | MEDLINE | ID: mdl-39155658

ABSTRACT

Objectives: Part of the tumor localization methods in radiotherapy have poor real-time performance and may generate additional radiation. We propose a multimodal point cloud-based method for tumor localization in robotic ultrasound-guided radiotherapy, which only irradiates computed tomography (CT) during radiotherapy planning to avoid additional radiation. Methods: The tumor position was determined using the CT point cloud, and the red green blue depth (RGBD) point cloud was used to determine body surface scanning location corresponding to the tumor location. The relationship between the CT point cloud and RGBD point cloud was established through multi-modal point cloud registration. The point cloud was then used for robot tumor localization through coordinate transformation between camera and robot. Results: The maximum mean absolute error of the tumor location in the X, Y, and Z directions of the robot coordinate system were 0.781, 1.334, and 1.490 mm, respectively. The average point-to-point translation mean absolute error between the actual and predicted positions of the localization points was 1.847 mm. The maximum error in the random positioning experiment was 1.77 mm. Conclusion: The proposed method is radiation free and has real-time performance, with tumor localization accuracy that meets the requirements of radiotherapy. The proposed method, which potentially reduces the risks associated with radiation exposure while ensuring efficient and accurate tumor localization, represents a promising advancement in the field of radiotherapy.


Subject(s)
Neoplasms , Radiotherapy Planning, Computer-Assisted , Radiotherapy, Image-Guided , Tomography, X-Ray Computed , Humans , Radiotherapy, Image-Guided/methods , Neoplasms/radiotherapy , Neoplasms/diagnostic imaging , Radiotherapy Planning, Computer-Assisted/methods , Tomography, X-Ray Computed/methods , Ultrasonography/methods , Algorithms , Phantoms, Imaging , Robotics/methods , Robotic Surgical Procedures/methods
5.
J Am Chem Soc ; 146(32): 22455-22468, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39094119

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) significantly hinder the immune response to tumor radiotherapy (RT) because of their massive accumulation in tumors after RT, resulting in immunosuppression and poor clinical prognosis. Herein, we developed an anti-PD-L1 antibody-conjugated iron oxide nanoprobe (Fe3O4-αPD-L1) to target and induce ferroptosis in MDSCs, thereby alleviating RT resistance. Overexpression of PD-L1 in MDSCs following RT enables noninvasive in vivo magnetic resonance and positron emission tomography imaging using 89Zr-labeled nanoprobes to track the movement of MDSCs and their infiltration into the tumor. After uptake by MDSCs that infiltrated the tumor, Fe3O4-αPD-L1 nanoprobes were mainly found within the lysosome and triggered the Fenton reaction, resulting in the generation of abundant reactive oxygen species. This process leads to ferroptosis of MDSCs, characterized by lipid peroxidation and mitochondrial dysfunction, and effectively reprograms the immunosuppressive environment within the tumor following RT. This study highlights a strategy for monitoring and regulating the fate of MDSCs to alleviate RT resistance and ultimately achieve improved treatment outcomes.


Subject(s)
Ferroptosis , Myeloid-Derived Suppressor Cells , Ferroptosis/drug effects , Myeloid-Derived Suppressor Cells/immunology , Animals , Humans , Mice , B7-H1 Antigen/metabolism , Neoplasms/diagnostic imaging , Neoplasms/radiotherapy , Neoplasms/pathology , Ferric Compounds/chemistry , Cell Line, Tumor
6.
J Am Chem Soc ; 146(32): 22689-22698, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39101919

ABSTRACT

Bioorthogonal pretargeting optical imaging shows the potential for enhanced diagnosis and prognosis. However, the bioorthogonal handles, known for being "always reactive", may engage in reactions at unintended sites with their counterparts, resulting in nonspecific fluorescence activation and diminishing detection specificity. Meanwhile, despite the importance of detecting senescent cancer cells in cancer therapy, current methods mainly rely on common single senescence-associated biomarkers, which lack specificity for differentiating between various types of senescent cells. Herein, we report a dual-locked enzyme-activatable bioorthogonal fluorescence (DEBOF) turn-on imaging approach for the specific detection of senescent cancer cells. A dual-locked bioorthogonal targeting agent (DBTA) and a bioorthogonally activatable fluorescent imaging probe (BAP) are synthesized as the biorthogonal pair. DBTA is a tetrazine derivative dually caged by two enzyme-cleavable moieties, respectively, associated with senescence and cancer, which ensures that its bioorthogonal reactivity ("clickability") is only triggered in the presence of senescent cancer cells. BAP is a fluorophore caged by trans-cyclooctane (TCO), whose fluorescence is only activated upon bioorthogonal reaction between its TCO and the decaged tetrazine of DBTA. As such, the DEBOF imaging approach differentiates senescent cancer cells from nonsenescent cancer cells or other senescent cells, allowing noninvasive tracking of the population fluctuation of senescent cancer cells in the tumor of living mice to guide cancer therapies. This study thus provides a general molecular strategy for biomarker-activatable in vivo bioorthogonal pretargeting imaging with the potential to be applied to other imaging modalities beyond optics.


Subject(s)
Cellular Senescence , Fluorescent Dyes , Optical Imaging , Humans , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Animals , Mice , Cell Line, Tumor , Neoplasms/diagnostic imaging , Fluorescence
7.
Nanotheranostics ; 8(4): 506-520, 2024.
Article in English | MEDLINE | ID: mdl-39135728

ABSTRACT

Photoacoustic imaging is a hybrid modality that combines high-contrast and spectroscopy-based optical imaging specificity with the high spatial resolution of ultrasonography. This review highlights the development and progress of photoacoustic imaging technology over the past decade. This imaging technology has evolved to be more user-friendly, cost-effective, and portable, demonstrating its potential for diverse clinical applications. A potential clinical application lies in the use of photoacoustic imaging as a guiding tool for photothermal therapy. This review was conducted by initially filtering through three databases, namely, Google Scholar, PubMed, and Scopus, resulting in 460 articles published between 2019 and May 2023. Of these, 54 articles were deemed suitable for review after identification. The selected articles were research papers focusing on the development of therapeutic agents that enhance contrast in photoacoustic imaging. All reviewed articles tested these agents both in vitro and in vivo. This review focuses on wavelength absorption and radiation sources for photothermal therapy. The developed agents predominantly used NIR-I wavelengths, whereas the NIR-II region has been less explored, indicating significant potential for future research. This review provides comprehensive insights into the advancement of compounds serving as therapeutic agents and contrast agents in photoacoustic imaging-guided photothermal therapy.


Subject(s)
Contrast Media , Photoacoustic Techniques , Photothermal Therapy , Photoacoustic Techniques/methods , Humans , Contrast Media/chemistry , Photothermal Therapy/methods , Animals , Neoplasms/therapy , Neoplasms/diagnostic imaging
8.
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
9.
Cancer Imaging ; 24(1): 107, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39148139

ABSTRACT

Extensive efforts have been dedicated to exploring the impact of tumor heterogeneity on cancer treatment at both histological and genetic levels. To accurately measure intra-tumoral heterogeneity, a non-invasive imaging technique, known as habitat imaging, was developed. The technique quantifies intra-tumoral heterogeneity by dividing complex tumors into distinct sub- regions, called habitats. This article reviews the following aspects of habitat imaging in cancer treatment, with a focus on radiotherapy: (1) Habitat imaging biomarkers for assessing tumor physiology; (2) Methods for habitat generation; (3) Efforts to combine radiomics, another imaging quantification method, with habitat imaging; (4) Technical challenges and potential solutions related to habitat imaging; (5) Pathological validation of habitat imaging and how it can be utilized to evaluate cancer treatment by predicting treatment response including survival rate, recurrence, and pathological response as well as ongoing open clinical trials.


Subject(s)
Magnetic Resonance Imaging , Neoplasms , Humans , Neoplasms/diagnostic imaging , Neoplasms/therapy , Magnetic Resonance Imaging/methods
10.
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
11.
J Photochem Photobiol B ; 258: 113000, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39121718

ABSTRACT

Lipid droplets (LDs) are spherical organelles that localize in the cytosol of eukaryotic cells. Different proteins are embedded on the surface of LDs, so LDs play a vital role in the physiological activities of cells. The dysregulation of LDs is associated with various human diseases, such as diabetes and obesity. Therefore, it is essential to develop a fluorescent dye that labels LDs to detect and monitor illnesses. In this study, we developed the compound BDAA12C for staining LDs in cells. BDAA12C exhibits excellent LD specificity and low toxicity, enabling us to successfully stain and observe the fusion of LDs in A549 cancer cells. Furthermore, we also successfully distinguished A549 cancer cells and MRC-5 normal cells in a co-culture experiment and in normal and tumour tissues. Interestingly, we found different localizations of BDAA12C in well-fed and starved A549 cancer cells and consequently illustrated the transfer of fatty acids (FAs) from LDs to mitochondria to supply energy for ß-oxidation upon starvation. Therefore, BDAA12C is a promising LD-targeted probe for cancer diagnosis and tracking lipid trafficking within cells.


Subject(s)
Fluorescent Dyes , Lipid Droplets , Humans , Lipid Droplets/metabolism , Lipid Droplets/chemistry , Fluorescent Dyes/chemistry , A549 Cells , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Fatty Acids/chemistry , Coculture Techniques , Mitochondria/metabolism , Acridines/chemistry , Microscopy, Fluorescence
12.
Nat Commun ; 15(1): 6931, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138215

ABSTRACT

Artificial intelligence (AI) algorithms hold the potential to revolutionize radiology. However, a significant portion of the published literature lacks transparency and reproducibility, which hampers sustained progress toward clinical translation. Although several reporting guidelines have been proposed, identifying practical means to address these issues remains challenging. Here, we show the potential of cloud-based infrastructure for implementing and sharing transparent and reproducible AI-based radiology pipelines. We demonstrate end-to-end reproducibility from retrieving cloud-hosted data, through data pre-processing, deep learning inference, and post-processing, to the analysis and reporting of the final results. We successfully implement two distinct use cases, starting from recent literature on AI-based biomarkers for cancer imaging. Using cloud-hosted data and computing, we confirm the findings of these studies and extend the validation to previously unseen data for one of the use cases. Furthermore, we provide the community with transparent and easy-to-extend examples of pipelines impactful for the broader oncology field. Our approach demonstrates the potential of cloud resources for implementing, sharing, and using reproducible and transparent AI pipelines, which can accelerate the translation into clinical solutions.


Subject(s)
Artificial Intelligence , Cloud Computing , Humans , Reproducibility of Results , Deep Learning , Radiology/methods , Radiology/standards , Algorithms , Neoplasms/diagnostic imaging , Image Processing, Computer-Assisted/methods
13.
Sci Adv ; 10(35): eadn8274, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39213344

ABSTRACT

Recent advancements in photoacoustic (PA) imaging have leveraged reversibly photoswitchable chromophores, known for their dual absorbance states, to enhance imaging sensitivity through differential techniques. Yet, their deployment in tumor imaging has faced obstacles in achieving targeted delivery with high efficiency and specificity. Addressing this challenge, we introduce innovative protein assemblies, DrBphP-CBD, by genetically fusing a photosensory module from Deinococcus radiodurans bacterial phytochrome (DrBphP) with a collagen-binding domain (CBD). These protein assemblies form sub-100-nanometer structures composed of 24 DrBphP dimers and 12 CBD trimers, presenting 24 protein subunits. Their affinity for collagens, combined with impressive photoswitching contrast, markedly improves PA imaging precision. In various tumor models, intravenous administration of DrBphP-CBD has demonstrated enhanced tumor targeting and retention, augmenting contrast in PA imaging by minimizing background noise. This strategy underscores the clinical potential of DrBphP-CBD as PA contrast agents, propelling photoswitchable chromoproteins to the forefront of precise cancer diagnosis.


Subject(s)
Collagen , Deinococcus , Neoplasms , Photoacoustic Techniques , Phytochrome , Photoacoustic Techniques/methods , Collagen/chemistry , Collagen/metabolism , Animals , Humans , Mice , Phytochrome/chemistry , Phytochrome/metabolism , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Deinococcus/metabolism , Deinococcus/chemistry , Cell Line, Tumor , Protein Binding , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/genetics
14.
Nat Commun ; 15(1): 7558, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39214974

ABSTRACT

Hydrogen sulfide is essential in numerous physiological and pathological processes and has emerged as a promising cancer imaging and signaling molecule and a potentially versatile therapeutic agent. However, the endogenous levels of hydrogen sulfide remain insufficient to perform its biological functions, and thus, developing novel strategies that amplify hydrogen sulfide signals at lesion sites is of increasing interest. In this work, a nanoplatform (SNP) based on hydrogen sulfide-responsive self-immolative poly(thiocarbamate) with localized hydrogen sulfide signal amplification capability is developed to encapsulate a hydrogen sulfide-responsive fluorescent probe (e.g., hemicyanine dye; p-Cy) or an anticancer prodrug (e.g., doxorubicin; p-DOX) to form a nanoprobe (SNPp-Cy) or nanomedicine (SNPp-DOX) for cancer imaging and therapy, respectively. SNPp-Cy exhibits a low detection limit for hydrogen sulfide, enabling ultrasensitive detection of small (<2 mm) tumors in female mice. In addition, SNPp-DOX can effectively inhibit the growth of DOX-resistant human breast cancer xenograft, lung metastasis, and patient-derived xenograft tumors in female mice.


Subject(s)
Doxorubicin , Hydrogen Sulfide , Hydrogen Sulfide/metabolism , Animals , Humans , Female , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Doxorubicin/administration & dosage , Mice , Cell Line, Tumor , Fluorescent Dyes/chemistry , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Mice, Nude , Xenograft Model Antitumor Assays , Prodrugs/pharmacology , Prodrugs/therapeutic use , Prodrugs/chemistry , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Mice, Inbred BALB C , Nanoparticles/chemistry , Optical Imaging/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
15.
J Nanobiotechnology ; 22(1): 515, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39198894

ABSTRACT

Recent advancements in cancer research have led to the generation of innovative nanomaterials for improved diagnostic and therapeutic strategies. Despite the proven potential of two-dimensional (2D) molybdenum disulfide (MoS2) as a versatile platform in biomedical applications, few review articles have focused on MoS2-based platforms for cancer theranostics. This review aims to fill this gap by providing a comprehensive overview of the latest developments in 2D MoS2 cancer theranostics and emerging strategies in this field. This review highlights the potential applications of 2D MoS2 in single-model imaging and therapy, including fluorescence imaging, photoacoustic imaging, photothermal therapy, and catalytic therapy. This review further classifies the potential of 2D MoS2 in multimodal imaging for diagnostic and synergistic theranostic platforms. In particular, this review underscores the progress of 2D MoS2 as an integrated drug delivery system, covering a broad spectrum of therapeutic strategies from chemotherapy and gene therapy to immunotherapy and photodynamic therapy. Finally, this review discusses the current challenges and future perspectives in meeting the diverse demands of advanced cancer diagnostic and theranostic applications.


Subject(s)
Disulfides , Molybdenum , Neoplasms , Theranostic Nanomedicine , Molybdenum/chemistry , Molybdenum/therapeutic use , Humans , Disulfides/chemistry , Theranostic Nanomedicine/methods , Neoplasms/therapy , Neoplasms/diagnostic imaging , Animals , Drug Delivery Systems/methods , Photoacoustic Techniques/methods , Nanostructures/chemistry , Nanostructures/therapeutic use , Multimodal Imaging/methods
16.
Talanta ; 279: 126629, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39106649

ABSTRACT

Considering the excellent properties such as deep tissue penetration, high signal-to-noise ratio, and in-situ recharge and reactivation, near-infrared luminescence long afterglow nanoparticles show considerable promise for biological application, especially in multifunctional imaging, targeting, and synergistic therapeutic. In this paper, Zn3Ga4GeO11: 0.1 % Cr3+, 1 % Yb3+, 0.1 % Tm3+@Ag-FA (ZGGO@Ag-FA, ZGA-FA) nanoparticles were synthesized by in-situ growth of Ag nanoparticles on the surface of long afterglow nanoparticles, and further modified with folic acid. Through precise adjustments, the luminescent properties of ZnGa2O4 were enhanced and notably boosted the photothermal effect of Ag by leveraging the upconversion emission of ZGGO, with a photothermal conversion efficiency reaching about 59.9 %. The ZGA-FA nanoparticles are ultra-small, measuring less than 50 nm. The modification with folic acid provides the ZGA-FA nanoparticles with excellent tumor-targeting capabilities, demonstrating effective enrichment and retention in tumor tissues, thus enabling long-term imaging and therapy through in vivo re-excitation. Due to its stable photothermal effect, outstanding near-infrared (NIR) afterglow imaging, and red-light charged characteristics, combined with effective tumor-targeting abilities, the therapeutic strategy proposed by this study has significant potential for clinical applications.


Subject(s)
Folic Acid , Animals , Humans , Mice , Folic Acid/chemistry , Optical Imaging , Silver/chemistry , Gallium/chemistry , Metal Nanoparticles/chemistry , Photothermal Therapy , Nanoparticles/chemistry , Mice, Inbred BALB C , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Neoplasms/therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Phototherapy , Mice, Nude
17.
Sci Rep ; 14(1): 18443, 2024 08 08.
Article in English | MEDLINE | ID: mdl-39117886

ABSTRACT

Hypoxia is an important feature of the tumor microenvironment (TME) of most solid tumors, and it is closely linked to cancer cell proliferation, therapy resistance, and the tumor immune response. Herein, we describe a method for hypoxia-induced heterogeneous oxygen distribution in xenograft tumors based on phosphorescence imaging microscopy (PLIM) using intravascular and intracellular oxygen probes. We synthesized Ir(III) complexes with polyethylene glycol (PEG) units of different molecular weights into the ligand as intravascular oxygen probes, BTP-PEGm (m = 2000, 5000, 10000, 20000). BTP-PEGm showed red emission with relatively high emission quantum yield and high oxygen sensitivity in saline. Cellular and in vivo experiments using these complexes revealed that BTP-PEG10000 was the most suitable probe in terms of blood retention and ease of intravenous administration in mice. PLIM measurements of xenograft tumors in mice treated with BTP-PEG10000 allowed simultaneous imaging of the tumor microvasculature and quantification of oxygen partial pressures. From lifetime images using the red-emitting intracellular oxygen probe BTPDM1 and the green-emitting intravascular fluorescent probe FITC-dextran, we demonstrated hypoxic heterogeneity in the TME with a sparse vascular network and showed that the oxygen levels of tumor cells gradually decreased with vascular distance.


Subject(s)
Microscopy, Confocal , Oxygen , Animals , Oxygen/metabolism , Mice , Humans , Microscopy, Confocal/methods , Cell Line, Tumor , Iridium/chemistry , Tumor Microenvironment , Polyethylene Glycols/chemistry , Mice, Nude , Fluorescent Dyes/chemistry , Neoplasms/diagnostic imaging , Neoplasms/pathology
18.
Nanoscale ; 16(33): 15585-15614, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39104307

ABSTRACT

Core-shell nanocomposites made of iron oxide core (IO NPs) coated with mesoporous silica (MS) shells are promising theranostic agents. While the core is being used as an efficient heating nanoagent under alternating magnetic field (AMF) and near infra-red (NIR) light and as a suitable contrast agent for magnetic resonance imaging (MRI), the MS shell is particularly relevant to ensure colloidal stability in a biological buffer and to transport a variety of therapeutics. However, a major challenge with such inorganic nanostructures is the design of adjustable silica structures, especially with tunable large pores which would be useful, for instance, for the delivery of large therapeutic biomolecule loading and further sustained release. Furthermore, the effect of tailoring a porous silica structure on the magneto- or photothermal dissipation still remains poorly investigated. In this work, we undertake an in-depth investigation of the growth of stellate mesoporous silica (STMS) shells around IO NPs cores and of their micro/mesoporous features respectively through time-lapse and in situ liquid phase transmission electron microscopy (LPTEM) and detailed nitrogen isotherm adsorption studies. We found here that the STMS shell features (thickness, pore size, surface area) can be finely tuned by simply controlling the sol-gel reaction time, affording a novel range of IO@STMS core@shell NPs. Finally, regarding the responses under alternating magnetic fields and NIR light which are evaluated as a function of the silica structure, IO@STMS NPs having a tunable silica shell structure are shown to be efficient as T2-weighted MRI agents and as heating agents for magneto- and photoinduced hyperthermia. Furthermore, such IO@STMS are found to display anti-cancer effects in pancreatic cancer cells under magnetic fields (both alternating and rotating).


Subject(s)
Ferric Compounds , Hyperthermia, Induced , Magnetic Resonance Imaging , Nanocomposites , Silicon Dioxide , Silicon Dioxide/chemistry , Nanocomposites/chemistry , Porosity , Humans , Ferric Compounds/chemistry , Cell Line, Tumor , Neoplasms/diagnostic imaging , Neoplasms/therapy , Neoplasms/drug therapy , Contrast Media/chemistry , Contrast Media/pharmacology
19.
Cell ; 187(17): 4458-4487, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39178829

ABSTRACT

Multiphoton fluorescence microscopy (MPFM) has been a game-changer for optical imaging, particularly for studying biological tissues deep within living organisms. MPFM overcomes the strong scattering of light in heterogeneous tissue by utilizing nonlinear excitation that confines fluorescence emission mostly to the microscope focal volume. This enables high-resolution imaging deep within intact tissue and has opened new avenues for structural and functional studies. MPFM has found widespread applications and has led to numerous scientific discoveries and insights into complex biological processes. Today, MPFM is an indispensable tool in many research communities. Its versatility and effectiveness make it a go-to technique for researchers investigating biological phenomena at the cellular and subcellular levels in their native environments. In this Review, the principles, implementations, capabilities, and limitations of MPFM are presented. Three application areas of MPFM, neuroscience, cancer biology, and immunology, are reviewed in detail and serve as examples for applying MPFM to biological research.


Subject(s)
Microscopy, Fluorescence, Multiphoton , Microscopy, Fluorescence, Multiphoton/methods , Humans , Animals , Neoplasms/diagnostic imaging , Neoplasms/pathology
20.
Spectrochim Acta A Mol Biomol Spectrosc ; 323: 124918, 2024 Dec 15.
Article in English | MEDLINE | ID: mdl-39096675

ABSTRACT

The higher viscosity and lower pH in lysosomes of cancer cells highlight their potential as biomarkers for cancer. Therefore, the development of acid-activated viscosity fluorescent probes is significant for the early diagnosis and treatment of cancer. Based on this, we have designed and synthesized a near-infrared fluorescent probe based on the 2-(2-hydroxyphenyl)benzothiazole (HBT) group, namely HBTH, to monitor the viscosity changes within lysosomes. It has been demonstrated that HBTH was extremely sensitive to viscosity, with a strong linear relationship between fluorescence intensity and log(viscosity) within the range of (logη) = 0-3.06 (a correlation coefficient of 0.98), proving its capability for quantitative viscosity measurement. In particular, the most obvious fluorescence enhancement of HBTH was only efficiently triggered by the combined effect of low pH and high viscosity. Furthermore, HBTH can rapidly localize to lysosomes by wash-free procedure at a low concentration (100 nM) and achieve high-fidelity imaging within 20 s. It can also monitor the dynamic processes of lysosomes in cells, viscosity changes under drug stimuli, and lysosomal behavior during mitophagy. Importantly, HBTH is capable of identifying tumors in tumor-bearing nude mice through in vivo imaging. These features make HBTH a powerful tool for the early diagnosis and treatment of cancer.


Subject(s)
Fluorescent Dyes , Lysosomes , Mice, Nude , Neoplasms , Lysosomes/metabolism , Lysosomes/chemistry , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Animals , Viscosity , Humans , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Neoplasms/pathology , Mice , Hydrogen-Ion Concentration , Cell Line, Tumor , Benzothiazoles/chemistry , Benzothiazoles/pharmacology , Mice, Inbred BALB C , Optical Imaging , Mitophagy/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL