Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 67
Filtrer
1.
Nanoscale Horiz ; 8(8): 1122, 2023 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-37382592

RÉSUMÉ

Correction for 'Multiplexed molecular imaging with surface enhanced resonance Raman scattering nanoprobes reveals immunotherapy response in mice via multichannel image segmentation' by Chrysafis Andreou et al., Nanoscale Horiz., 2022, 7, 1540-1552, https://doi.org/10.1039/d2nh00331g.

2.
ACS Nano ; 17(5): 4261-4278, 2023 03 14.
Article de Anglais | MEDLINE | ID: mdl-36706095

RÉSUMÉ

Triple-negative breast cancer (TNBC) is considered more aggressive with a poorer prognosis than other breast cancer subtypes. Through systemic bioinformatic analyses, we established the ferroptosis potential index (FPI) based on the expression profile of ferroptosis regulatory genes and found that TNBC has a higher FPI than non-TNBC in human BC cell lines and tumor tissues. To exploit this finding for potential patient stratification, we developed biologically amenable phototheranostic iron pyrite FeS2 nanocrystals (NCs) that efficiently harness near-infrared (NIR) light, as in photovoltaics, for multispectral optoacoustic tomography (MSOT) and photothermal ablation with a high photothermal conversion efficiency (PCE) of 63.1%. Upon NIR irradiation that thermodynamically enhances Fenton reactions, dual death pathways of apoptosis and ferroptosis are simultaneously triggered in TNBC cells, comprehensively limiting primary and metastatic TNBC by regulating p53, FoxO, and HIF-1 signaling pathways and attenuating a series of metabolic processes, including glutathione and amino acids. As a unitary phototheranostic agent with a safe toxicological profile, the nanocrystal represents an effective way to circumvent the lack of therapeutic targets and the propensity of multisite metastatic progression in TNBC in a streamlined workflow of cancer management with an integrated image-guided intervention.


Sujet(s)
Nanoparticules , Photosensibilisants , Thérapie photothermique , Tumeurs du sein triple-négatives , Humains , Mort cellulaire , Lignée cellulaire tumorale , Fer/administration et posologie , Fer/usage thérapeutique , Nanoparticules/administration et posologie , Nanoparticules/usage thérapeutique , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/radiothérapie , Femelle , Rayons infrarouges/usage thérapeutique , Thérapie photothermique/méthodes , Sulfures/administration et posologie , Sulfures/usage thérapeutique , Photosensibilisants/administration et posologie , Photosensibilisants/usage thérapeutique , Apoptose/effets des médicaments et des substances chimiques , Apoptose/effets des radiations , Ferroptose/effets des médicaments et des substances chimiques , Ferroptose/effets des radiations
3.
Nanoscale Horiz ; 7(12): 1540-1552, 2022 11 21.
Article de Anglais | MEDLINE | ID: mdl-36285605

RÉSUMÉ

Visualizing the presence and distribution of multiple specific molecular markers within a tumor can reveal the composition of its microenvironment, inform diagnosis, stratify patients, and guide treatment. Raman imaging with multiple molecularly-targeted surface enhanced Raman scattering (SERS) nanoprobes could help investigate emerging cancer treatments preclinically or enable personalized treatment assessment. Here, we report a comprehensive strategy for multiplexed imaging using SERS nanoprobes and machine learning (ML) to monitor the early effects of immune checkpoint blockade (ICB) in tumor-bearing mice. We used antibody-functionalized SERS nanoprobes to visualize 7 + 1 immunotherapy-related targets simultaneously. The multiplexed images were spectrally resolved and then spatially segmented into superpixels based on the unmixed signals. The superpixels were used to train ML models, leading to the successful classification of mice into treated and untreated groups, and identifying tumor regions with variable responses to treatment. This method may help predict treatment efficacy in tumors and identify areas of tumor variability and therapy resistance.


Sujet(s)
Tumeurs , Analyse spectrale Raman , Souris , Animaux , Analyse spectrale Raman/méthodes , Immunothérapie , Anticorps/usage thérapeutique , Tumeurs/imagerie diagnostique , Tumeurs/thérapie , Facteurs immunologiques , Imagerie moléculaire , Microenvironnement tumoral
4.
Nat Biomed Eng ; 6(5): 527-540, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-35624151

RÉSUMÉ

In oncology, technologies for clinical molecular imaging are used to diagnose patients, establish the efficacy of treatments and monitor the recurrence of disease. Multiplexed methods increase the number of disease-specific biomarkers that can be detected simultaneously, such as the overexpression of oncogenic proteins, aberrant metabolite uptake and anomalous blood perfusion. The quantitative localization of each biomarker could considerably increase the specificity and the accuracy of technologies for clinical molecular imaging to facilitate granular diagnoses, patient stratification and earlier assessments of the responses to administered therapeutics. In this Review, we discuss established techniques for multiplexed imaging and the most promising emerging multiplexing technologies applied to the imaging of isolated tissues and cells and to non-invasive whole-body imaging. We also highlight advances in radiology that have been made possible by multiplexed imaging.


Sujet(s)
Imagerie diagnostique , Marqueurs biologiques , Humains
5.
Nanotheranostics ; 6(3): 256-269, 2022.
Article de Anglais | MEDLINE | ID: mdl-35145836

RÉSUMÉ

Cell surface marker expression in tumors dictates the selection of therapeutics, therapy response, and survival. However, biopsies are invasive, sample only a small area of the tumor landscape and may miss significant areas of heterogeneous expression. Here, we investigated the potential of antibody-conjugated surface-enhanced resonance Raman scattering nanoparticles (SERRS-NPs) to depict and quantify high and low tumoral surface marker expression, focusing on the surface markers epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2) in an intracerebral and peripheral setting with an inter- and intratumoral comparison of Raman signal intensities. Methods: ICR-Prkdc mice were injected with glioblastoma, epidermoid carcinoma, or breast tumor cell lines intracerebrally and peripherally. SERRS-NPs were functionalized with cetuximab or trastuzumab and administered via tail vein injection. Raman imaging was performed 18 hours post-injection in excised tumors and in vivo through the skull. Tumors were then fixed and processed for immunohistochemical evaluation. Results: Confirmed by MRI and immunohistochemistry for EGFR and HER2, our results demonstrate that antibody-conjugated SERRS-NPs go beyond the delineation of a tumor and offer clear and distinct Raman spectra that reflect the distribution of the targeted surface marker. The intensity of the SERRS-NP signal accurately discriminated high- versus low-expressing surface markers between tumors, and between different areas within tumors. Conclusion: Biopsies can be highly invasive procedures and provide a limited sample of molecular expression within a tumor. Our nanoparticle-based Raman imaging approach offers the potential to provide non-invasive and more comprehensive molecular imaging and an alternative to the current clinical gold standard of immunohistochemistry.


Sujet(s)
Glioblastome , Nanoparticules , Animaux , Modèles animaux de maladie humaine , Glioblastome/imagerie diagnostique , Glioblastome/anatomopathologie , Souris , Souris de lignée ICR , Nanoparticules/composition chimique , Analyse spectrale Raman/méthodes
6.
Nanotheranostics ; 6(1): 10-30, 2022.
Article de Anglais | MEDLINE | ID: mdl-34976578

RÉSUMÉ

Surface-enhanced Raman spectroscopy (SERS) nanotags hold a unique place among bioimaging contrast agents due to their fingerprint-like spectra, which provide one of the highest degrees of detection specificity. However, in order to achieve a sufficiently high signal intensity, targeting capabilities, and biocompatibility, all components of nanotags must be rationally designed and tailored to a specific application. Design parameters include fine-tuning the properties of the plasmonic core as well as optimizing the choice of Raman reporter molecule, surface coating, and targeting moieties for the intended application. This review introduces readers to the principles of SERS nanotag design and discusses both established and emerging protocols of their synthesis, with a specific focus on the construction of SERS nanotags in the context of bioimaging and theranostics.


Sujet(s)
Or , Analyse spectrale Raman , Analyse spectrale Raman/méthodes
7.
Theranostics ; 11(6): 2534-2549, 2021.
Article de Anglais | MEDLINE | ID: mdl-33456558

RÉSUMÉ

Rationale: Most contemporary cancer therapeutic paradigms involve initial imaging as a treatment roadmap, followed by the active engagement of surgical operations. Current approved intraoperative contrast agents exemplified by indocyanine green (ICG) have a few drawbacks including the inability of pre-surgical localization. Alternative near-infrared (NIR) dyes including IRDye800cw are being explored in advanced clinical trials but often encounter low chemical yields and complex purifications owing to the asymmetric synthesis. A single contrast agent with ease of synthesis that works in multiple cancer types and simultaneously allows presurgical imaging, intraoperative deep-tissue three-dimensional visualization, and high-speed microscopic visualization of tumor margins via spatiotemporally complementary modalities would be beneficial. Methods: Due to the lack of commercial availability and the absence of detailed synthesis and characterization, we proposed a facile and scalable synthesis pathway for the symmetric NIR water-soluble heptamethine sulfoindocyanine IRDye78. The synthesis can be accomplished in four steps from commercially-available building blocks. Its symmetric resonant structure avoided asymmetric synthesis problems while still preserving the benefits of analogous IRDye800cw with commensurable optical properties. Next, we introduced a low-molecular-weight protein alpha-lactalbumin (α-LA) as the carrier that effectively modulates the hepatic clearance of IRDye78 into the preferred renal excretion pathway. We further implemented 89Zr radiolabeling onto the protein scaffold for positron emission tomography (PET). The multimodal imaging capability of the fluorophore-protein complex was validated in breast cancer and glioblastoma. Results: The scalable synthesis resulted in high chemical yields, typically 95% yield in the final step of the chloro dye. Chemical structures of intermediates and the final fluorophore were confirmed. Asymmetric IRDye78 exhibited comparable optical features as symmetric IRDye800cw. Its well-balanced quantum yield affords concurrent dual fluorescence and optoacoustic contrast without self-quenching nor concentration-dependent absorption. The NHS ester functionality modulates efficient covalent coupling to reactive side-chain amines to the protein carrier, along with desferrioxamine (DFO) for stable radiolabeling of 89Zr. The fluorophore-protein complex advantageously shifted the biodistribution and can be effectively cleared through the urinary pathway. The agent accumulates in tumors and enables triple-modal visualization in mouse xenograft models of both breast and brain cancers. Conclusion: This study described in detail a generalized strategic modulation of clearance routes towards the favorable renal clearance, via the introduction of α-LA. IRDye78 as a feasible alternative of IRDye800cw currently in clinical phases was proposed with a facile synthesis and fully characterized for the first time. This fluorophore-protein complex with stable radiolabeling should have great potential for clinical translation where it could enable an elegant workflow from preoperative planning to intraoperative deep tissue and high-resolution image-guided resection.


Sujet(s)
Tumeurs du cerveau/imagerie diagnostique , Colorants fluorescents/métabolisme , Glioblastome/imagerie diagnostique , Vert indocyanine/métabolisme , Imagerie optique/méthodes , Spectroscopie proche infrarouge/méthodes , Animaux , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/chirurgie , Lignée cellulaire tumorale , Femelle , Fluorescence , Glioblastome/métabolisme , Glioblastome/chirurgie , Humains , Indoles/métabolisme , Lactalbumine/métabolisme , Souris , Souris de lignée C57BL , Tomographie par émission de positons/méthodes , Distribution tissulaire , Tomodensitométrie/méthodes
8.
Nanotheranostics ; 5(1): 90-112, 2021.
Article de Anglais | MEDLINE | ID: mdl-33391977

RÉSUMÉ

Immunotherapy has revolutionized the treatment of several malignancies. Notwithstanding the encouraging results, many patients do not respond to treatments. Evaluation of the efficacy of treatments is challenging and robust methods to predict the response to treatment are not yet available. The outcome of immunotherapy results from changes that treatment evokes in the tumor immune landscape. Therefore, a better understanding of the dynamics of immune cells that infiltrate into the tumor microenvironment may fundamentally help in addressing this challenge and provide tools to assess or even predict the response. Noninvasive imaging approaches, such as PET and SPECT that provide whole-body images are currently seen as the most promising tools that can shed light on the events happening in tumors in response to treatment. Such tools can provide critical information that can be used to make informed clinical decisions. Here, we review recent developments in the field of noninvasive cancer imaging with a focus on immunotherapeutics and nuclear imaging technologies and will discuss how the field can move forward to address the challenges that remain unresolved.


Sujet(s)
Immunothérapie , Tumeurs/thérapie , Humains , Microenvironnement tumoral
9.
Chem Soc Rev ; 50(1): 556-568, 2021 Jan 07.
Article de Anglais | MEDLINE | ID: mdl-33169761

RÉSUMÉ

In recent years, Raman spectroscopy has undergone major advancements in its ability to probe deeply through turbid media such as biological tissues. This progress has been facilitated by the advent of a range of specialist techniques based around spatially offset Raman spectroscopy (SORS) to enable non-invasive probing of living tissue through depths of up to 5 cm. This represents an improvement in depth penetration of up to two orders of magnitude compared to what can be achieved with conventional Raman methods. In combination with the inherently high molecular specificity of Raman spectroscopy, this has therefore opened up entirely new prospects for a range of new analytical applications across multiple fields including medical diagnosis and disease monitoring. This article discusses SORS and related variants of deep Raman spectroscopy such as transmission Raman spectroscopy (TRS), micro-SORS and surface enhanced spatially offset Raman spectroscopy (SESORS), and reviews the progress made in this field during the past 5 years including advances in non-invasive cancer diagnosis, monitoring of neurotransmitters, and assessment of bone disease.


Sujet(s)
Recherche biomédicale , Maladies osseuses/diagnostic , Tumeurs/diagnostic , Agents neuromédiateurs/analyse , Animaux , Humains , Analyse spectrale Raman
10.
Adv Sci (Weinh) ; 7(23): 2001669, 2020 Dec.
Article de Anglais | MEDLINE | ID: mdl-33304747

RÉSUMÉ

In the last two decades, DNA has attracted significant attention toward the development of materials at the nanoscale for emerging applications due to the unparalleled versatility and programmability of DNA building blocks. DNA-based artificial nanomaterials can be broadly classified into two categories: DNA nanostructures (DNA-NSs) and DNA-functionalized nanoparticles (DNA-NPs). More importantly, their use in nanotheranostics, a field that combines diagnostics with therapy via drug or gene delivery in an all-in-one platform, has been applied extensively in recent years to provide personalized cancer treatments. Conveniently, the ease of attachment of both imaging and therapeutic moieties to DNA-NSs or DNA-NPs enables high biostability, biocompatibility, and drug loading capabilities, and as a consequence, has markedly catalyzed the rapid growth of this field. This review aims to provide an overview of the recent progress of DNA-NSs and DNA-NPs as theranostic agents, the use of DNA-NSs and DNA-NPs as gene and drug delivery platforms, and a perspective on their clinical translation in the realm of oncology.

11.
Nat Biomed Eng ; 4(7): 686-703, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32661307

RÉSUMÉ

Theranostic agents should ideally be renally cleared and biodegradable. Here, we report the synthesis, characterization and theranostic applications of fluorescent ultrasmall gold quantum clusters that are stabilized by the milk metalloprotein alpha-lactalbumin. We synthesized three types of these nanoprobes that together display fluorescence across the visible and near-infrared spectra when excited at a single wavelength through optical colour coding. In live tumour-bearing mice, the near-infrared nanoprobe generates contrast for fluorescence, X-ray computed tomography and magnetic resonance imaging, and exhibits long circulation times, low accumulation in the reticuloendothelial system, sustained tumour retention, insignificant toxicity and renal clearance. An intravenously administrated near-infrared nanoprobe with a large Stokes shift facilitated the detection and image-guided resection of breast tumours in vivo using a smartphone with modified optics. Moreover, the partially unfolded structure of alpha-lactalbumin in the nanoprobe helps with the formation of an anti-cancer lipoprotein complex with oleic acid that triggers the inhibition of the MAPK and PI3K-AKT pathways, immunogenic cell death and the recruitment of infiltrating macrophages. The biodegradability and safety profile of the nanoprobes make them suitable for the systemic detection and localized treatment of cancer.


Sujet(s)
Tumeurs du sein/imagerie diagnostique , Tumeurs du sein/traitement médicamenteux , Or/composition chimique , Or/pharmacologie , Lactalbumine/composition chimique , Lactalbumine/pharmacologie , Animaux , Apoptose , Tumeurs du sein/anatomopathologie , Mort cellulaire , Femelle , Hétérogreffes , Lipoprotéines , Imagerie par résonance magnétique/méthodes , Souris , Souris de lignée BALB C , Mitogen-Activated Protein Kinase Kinases/effets des médicaments et des substances chimiques , Nanotechnologie/méthodes , Imagerie optique , Phosphatidylinositol 3-kinases/effets des médicaments et des substances chimiques , Protéomique , Nanomédecine théranostique/méthodes
12.
Chemistry ; 25(64): 14517-14521, 2019 Nov 18.
Article de Anglais | MEDLINE | ID: mdl-31515825

RÉSUMÉ

Distortion of nominally planar phthalocyanine macrocycles affects the excited state dynamics in that most of the excited-state energy decays through internal conversion. A click-type annulation reaction on a perfluorophthalocyanine platform appending a seven-membered ring to the ß-positions on one or more of the isoindoles distorts the macrocycle and modulates solubility. The distorted derivative enables photoacoustic imaging, photothermal effects, and strong surface-enhanced resonance Raman signals.

13.
Theranostics ; 9(20): 5899-5913, 2019.
Article de Anglais | MEDLINE | ID: mdl-31534527

RÉSUMÉ

Rationale: The goal of imaging tumors at depth with high sensitivity and specificity represents a significant challenge in the field of biomedical optical imaging. 'Surface enhanced Raman scattering' (SERS) nanoparticles (NPs) have been employed as image contrast agents and can be used to specifically target cells in vivo. By tracking their unique "fingerprint" spectra, it becomes possible to determine their precise location. However, while the detection of SERS NPs is very sensitive and specific, conventional Raman spectroscopy imaging devices are limited in their inability to probe through tissue depths of more than a few millimetres, due to scattering and absorption of photons by biological tissues. Here, we combine the use of "Spatially Offset Raman spectroscopy" (SORS) with that of "surface-enhanced resonance Raman spectroscopy" (SERRS) in a technique known as "surface enhanced spatially offset resonance Raman spectroscopy" (SESO(R)RS) to image deep-seated glioblastoma multiforme (GBM) tumors in vivo in mice through the intact skull. Methods: A SORS imaging system was built in-house. Proof of concept SORS imaging was achieved using a PTFE-skull-tissue phantom. Imaging of GBMs in the RCAS-PDGF/N-tva transgenic mouse model was achieved through the use of gold nanostars functionalized with a resonant Raman reporter to create SERRS nanostars. These were then encapsulated in a thin silica shell and functionalized with a cyclic-RGDyK peptide to yield integrin-targeting SERRS nanostars. Non-invasive in vivo SORS image acquisition of the integrin-targeted nanostars was then performed in living mice under general anesthesia. Conventional non-SORS imaging was used as a direct comparison. Results: Using a low power density laser, GBMs were imaged via SESORRS in mice (n = 5) and confirmed using MRI and histopathology. The results demonstrate that via utilization of the SORS approach, it is possible to acquire clear and distinct Raman spectra from deep-seated GBMs in mice in vivo through the skull. SESORRS images generated using classical least squares outlined the tumors with high precision as confirmed via MRI and histology. Unlike SESORRS, conventional Raman imaging of the same areas did not provide a clear delineation of the tumor. Conclusion: To the best of our knowledge this is the first report of in vivo SESO(R)RS imaging. In a relevant brain tumor mouse model we demonstrate that this technique can overcome the limitations of conventional Raman imaging with regards to penetration depth. This work therefore represents a significant step forward in the potential clinical translation of SERRS nanoparticles for high precision cancer imaging.


Sujet(s)
Glioblastome/imagerie diagnostique , Or/composition chimique , Nanoparticules métalliques/composition chimique , Analyse spectrale Raman/méthodes , Animaux , Tumeurs du cerveau/imagerie diagnostique , Modèles animaux de maladie humaine , Souris , Oligopeptides/composition chimique
14.
Sci Rep ; 9(1): 8550, 2019 06 12.
Article de Anglais | MEDLINE | ID: mdl-31189972

RÉSUMÉ

Breast cancer is the most common type of malignant growth in women. Early detection of breast cancer, as well as the identification of possible metastatic spread poses a significant challenge because of the structural and genetic heterogeneity that occurs during the progression of the disease. Currently, mammographies, biopsies and MRI scans are the standard of care techniques used for breast cancer diagnosis, all of which have their individual shortfalls, especially when it comes to discriminating tumors and benign growths. With this in mind, we have developed a non-invasive optoacoustic imaging strategy that targets the acidic environment of breast cancer. A pH low insertion peptide (pHLIP) was conjugated to the dark quencher QC1, yielding a non-fluorescent sonophore with high extinction coefficient in the near infrared that increases signal as a function of increasing amounts of membrane insertion. In an orthotopic murine breast cancer model, pHLIP-targeted optoacoustic imaging allowed us to differentiate between healthy and breast cancer tissues with high signal/noise ratios. In vivo, the sonophore QC1-pHLIP could detect malignancies at higher contrast than its fluorescent analog ICG-pHLIP, which was developed for fluorescence-guided surgical applications. PHLIP-type optoacoustic imaging agents in clinical settings are attractive due to their ability to target breast cancer and a wide variety of other malignant growths for diagnostic purposes. Intuitively, these agents could also be used for visualization during surgery.


Sujet(s)
Produits de contraste , Tumeurs expérimentales de la mamelle/imagerie diagnostique , Techniques photoacoustiques , Animaux , Lignée cellulaire tumorale , Produits de contraste/synthèse chimique , Produits de contraste/composition chimique , Produits de contraste/pharmacologie , Femelle , Tumeurs expérimentales de la mamelle/métabolisme , Tumeurs expérimentales de la mamelle/anatomopathologie , Souris , Souris transgéniques
15.
J Vis Exp ; (145)2019 03 25.
Article de Anglais | MEDLINE | ID: mdl-30958459

RÉSUMÉ

Ovarian cancer represents the deadliest gynecologic malignancy. Most patients present at an advanced stage (FIGO stage III or IV), when local metastatic spread has already occurred. However, ovarian cancer has a unique pattern of metastatic spread, in that tumor implants are initially contained within the peritoneal cavity. This feature could enable, in principle, the complete resection of tumor implants with curative intent. Many of these metastatic lesions are microscopic, making them hard to identify and treat. Neutralizing such micrometastases is believed to be a major goal towards eliminating tumor recurrence and achieving long-term survival. Raman imaging with surface enhanced resonance Raman scattering nanoprobes can be used to delineate microscopic tumors with high sensitivity, due to their bright and bioorthogonal spectral signatures. Here, we describe the synthesis of two 'flavors' of such nanoprobes: an antibody-functionalized one that targets the folate receptor - overexpressed in many ovarian cancers - and a non-targeted control nanoprobe, with distinct spectra. The nanoprobes are co-administered intraperitoneally to mouse models of metastatic human ovarian adenocarcinoma. All animal studies were approved by the Institutional Animal Care and Use Committee of Memorial Sloan Kettering Cancer Center. The peritoneal cavity of the animals is surgically exposed, washed, and scanned with a Raman microphotospectrometer. Subsequently, the Raman signatures of the two nanoprobes are decoupled using a Classical Least Squares fitting algorithm, and their respective scores divided to provide a ratiometric signal of folate-targeted over untargeted probes. In this way, microscopic metastases are visualized with high specificity. The main benefit of this approach is that the local application into the peritoneal cavity - which can be done conveniently during the surgical procedure - can tag tumors without subjecting the patient to systemic nanoparticle exposure. False positive signals stemming from non-specific binding of the nanoprobes onto visceral surfaces can be eliminated by following a ratiometric approach where targeted and non-targeted nanoprobes with distinct Raman signatures are applied as a mixture. The procedure is currently still limited by the lack of a commercial wide-field Raman imaging camera system, which once available will allow for the application of this technique in the operating theater.


Sujet(s)
Récepteurs des folates à ancre GPI/métabolisme , Nanotechnologie/méthodes , Tumeurs de l'ovaire/diagnostic , Analyse spectrale Raman/méthodes , Animaux , Lignée cellulaire tumorale , Femelle , Humains , Souris , Tumeurs de l'ovaire/métabolisme , Tumeurs de l'ovaire/anatomopathologie , Récidive , Sensibilité et spécificité
16.
Nat Commun ; 10(1): 1926, 2019 04 26.
Article de Anglais | MEDLINE | ID: mdl-31028250

RÉSUMÉ

Recently, surface-enhanced Raman scattering nanoprobes have shown tremendous potential in oncological imaging owing to the high sensitivity and specificity of their fingerprint-like spectra. As current Raman scanners rely on a slow, point-by-point spectrum acquisition, there is an unmet need for faster imaging to cover a clinically relevant area in real-time. Herein, we report the rational design and optimization of fluorescence-Raman bimodal nanoparticles (FRNPs) that synergistically combine the specificity of Raman spectroscopy with the versatility and speed of fluorescence imaging. DNA-enabled molecular engineering allows the rational design of FRNPs with a detection limit as low as 5 × 10-15 M. FRNPs selectively accumulate in tumor tissue mouse cancer models and enable real-time fluorescence imaging for tumor detection, resection, and subsequent Raman-based verification of clean margins. Furthermore, FRNPs enable highly efficient image-guided photothermal ablation of tumors, widening the scope of the NPs into the therapeutic realm.


Sujet(s)
Tumeurs du cerveau/thérapie , ADN/composition chimique , Nanoparticules métalliques/composition chimique , Imagerie optique/méthodes , Tumeurs de l'ovaire/thérapie , Analyse spectrale Raman/méthodes , Animaux , Tumeurs du cerveau/imagerie diagnostique , Tumeurs du cerveau/génétique , Tumeurs du cerveau/chirurgie , Lignée cellulaire tumorale , ADN/métabolisme , Vecteurs de médicaments/synthèse chimique , Vecteurs de médicaments/pharmacocinétique , Femelle , Colorants fluorescents/composition chimique , Génie génétique , Humains , Thérapie laser/instrumentation , Thérapie laser/méthodes , Limite de détection , Photothérapie de faible intensité/instrumentation , Photothérapie de faible intensité/méthodes , Nanoparticules métalliques/administration et posologie , Souris , Imagerie optique/instrumentation , Tumeurs de l'ovaire/imagerie diagnostique , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/chirurgie , Fantômes en imagerie , Argent/composition chimique , Analyse spectrale Raman/instrumentation , Tests d'activité antitumorale sur modèle de xénogreffe
17.
ACS Nano ; 13(2): 1354-1364, 2019 02 26.
Article de Anglais | MEDLINE | ID: mdl-30624916

RÉSUMÉ

Cancers of the gastrointestinal (GI) tract are among the most frequent and most lethal cancers worldwide. An important reason for this high mortality is that early disease is typically asymptomatic, and patients often present with advanced, incurable disease. Even in high-risk patients who routinely undergo endoscopic screening, lesions can be missed due to their small size or subtle appearance. Thus, current imaging approaches lack the sensitivity and specificity to accurately detect incipient GI tract cancers. Here we report our finding that a single dose of a high-sensitivity surface-enhanced resonance Raman scattering nanoparticle (SERRS-NP) enables reliable detection of precancerous GI lesions in animal models that closely mimic disease development in humans. Some of these animal models have not been used previously to evaluate imaging probes for early cancer detection. The studies were performed using a commercial Raman imaging system, a newly developed mouse Raman endoscope, and finally a clinically applicable Raman endoscope for larger animal studies. We show that this SERRS-NP-based approach enables robust detection of small, premalignant lesions in animal models that faithfully recapitulate human esophageal, gastric, and colorectal tumorigenesis. This method holds promise for much earlier detection of GI cancers than currently possible and could lead therefore to marked reduction of morbidity and mortality of these tumor types.


Sujet(s)
Endoscopie/méthodes , Tumeurs gastro-intestinales/diagnostic , Tube digestif/métabolisme , Nanoparticules/composition chimique , Analyse spectrale Raman/méthodes , Animaux , Modèles animaux de maladie humaine , Mâle , Souris , Souris de lignée C57BL , Souris knockout
18.
Nanotheranostics ; 2(3): 269-270, 2018.
Article de Anglais | MEDLINE | ID: mdl-29911001
19.
Small ; 14(23): e1800740, 2018 06.
Article de Anglais | MEDLINE | ID: mdl-29726109

RÉSUMÉ

Difficulty in visualizing glioma margins intraoperatively remains a major issue in the achievement of gross total tumor resection and, thus, better clinical outcome of glioblastoma (GBM) patients. Here, the potential of a new combined optical + optoacoustic imaging method for intraoperative brain tumor delineation is investigated. A strategy using a newly developed gold nanostar synthesis method, Raman reporter chemistry, and silication method to produce dual-modality contrast agents for combined surface-enhanced resonance Raman scattering (SERRS) and multispectral optoacoustic tomography (MSOT) imaging is devised. Following intravenous injection of the SERRS-MSOT-nanostars in brain tumor bearing mice, sequential MSOT imaging is performed in vivo and followed by Raman imaging. MSOT is able to accurately depict GBMs three-dimensionally with high specificity. The MSOT signal is found to correlate well with the SERRS images. Because SERRS enables uniquely sensitive high-resolution surface detection, it could represent an ideal complementary imaging modality to MSOT, which enables real-time, deep tissue imaging in 3D. This dual-modality SERRS-MSOT-nanostar contrast agent reported here is shown to enable high precision depiction of the extent of infiltrating GBMs by Raman- and MSOT imaging in a clinically relevant murine GBM model and could pave new ways for improved image-guided resection of brain tumors.


Sujet(s)
Tumeurs du cerveau/diagnostic , Nanoparticules/composition chimique , Techniques photoacoustiques/méthodes , Analyse spectrale Raman/méthodes , Tomographie/méthodes , Animaux , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/ultrastructure , Glioblastome/diagnostic , Glioblastome/anatomopathologie , Glioblastome/ultrastructure , Humains , Souris
20.
J Neurooncol ; 139(1): 125-133, 2018 Aug.
Article de Anglais | MEDLINE | ID: mdl-29619649

RÉSUMÉ

INTRODUCTION: The aim of the present study is to assess whether postoperative residual non-enhancing volume (PRNV) is correlated and predictive of overall survival (OS) in glioblastoma (GBM) patients. METHODS: We retrospectively analyzed a total 134 GBM patients obtained from The University of Texas MD Anderson Cancer Center (training cohort, n = 97) and The Cancer Genome Atlas (validation cohort, n = 37). All patients had undergone postoperative magnetic resonance imaging immediately after surgery. We evaluated the survival outcomes with regard to PRNV. The role of possible prognostic factors that may affect survival after resection, including age, sex, preoperative Karnofsky performance status, postoperative nodular enhancement, surgically induced enhancement, and postoperative necrosis, was investigated using univariate and multivariate Cox proportional hazards regression analyses. Additionally, a recursive partitioning analysis (RPA) was used to identify prognostic groups. RESULTS: Our analyses revealed that a high PRNV (HR 1.051; p-corrected = 0.046) and old age (HR 1.031; p-corrected = 0.006) were independent predictors of overall survival. This trend was also observed in the validation cohort (higher PRNV: HR 1.127, p-corrected = 0.002; older age: HR 1.034, p-corrected = 0.022). RPA analysis identified two prognostic risk groups: low-risk group (PRNV < 70.2 cm3; n = 55) and high-risk group (PRNV ≥ 70.2 cm3; n = 42). GBM patients with low PRNV had a significant survival benefit (5.6 months; p = 0.0037). CONCLUSION: Our results demonstrate that high PRNV is associated with poor OS. Such results could be of great importance in a clinical setting, particularly in the postoperative management and monitoring of therapy.


Sujet(s)
Tumeurs du cerveau/imagerie diagnostique , Encéphale/imagerie diagnostique , Glioblastome/imagerie diagnostique , Imagerie par résonance magnétique , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Encéphale/chirurgie , Tumeurs du cerveau/mortalité , Tumeurs du cerveau/chirurgie , Femelle , Glioblastome/mortalité , Glioblastome/chirurgie , Humains , Interprétation d'images assistée par ordinateur , Mâle , Adulte d'âge moyen , Période postopératoire , Pronostic , Études rétrospectives , Jeune adulte
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE