Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Nanomedicine ; 54: 102711, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37813236

ABSTRACT

For the past decades, gene editing demonstrated the potential to attenuate each of the root causes of genetic, infectious, immune, cancerous, and degenerative disorders. More recently, Clustered Regularly Interspaced Short Palindromic Repeats-CRISPR-associated protein 9 (CRISPR-Cas9) editing proved effective for editing genomic, cancerous, or microbial DNA to limit disease onset or spread. However, the strategies to deliver CRISPR-Cas9 cargos and elicit protective immune responses requires safe delivery to disease targeted cells and tissues. While viral vector-based systems and viral particles demonstrate high efficiency and stable transgene expression, each are limited in their packaging capacities and secondary untoward immune responses. In contrast, the nonviral vector lipid nanoparticles were successfully used for as vaccine and therapeutic deliverables. Herein, we highlight each available gene delivery systems for treating and preventing a broad range of infectious, inflammatory, genetic, and degenerative diseases. STATEMENT OF SIGNIFICANCE: CRISPR-Cas9 gene editing for disease treatment and prevention is an emerging field that can change the outcome of many chronic debilitating disorders.


Subject(s)
CRISPR-Cas Systems , Gene Editing , CRISPR-Cas Systems/genetics , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Gene Transfer Techniques , Genetic Therapy
2.
J Photochem Photobiol B ; 242: 112683, 2023 May.
Article in English | MEDLINE | ID: mdl-36934549

ABSTRACT

The primary treatment for malignant tumors remains to be surgical removal of the diseased tissue. The presence or absence of residual diseased tissue at the tumor margin is the strongest predictor of postoperative prognosis and recurrence. Accordingly, reliance on the ability of surgeons to visually distinguish diseased tissue from healthy tissue unambiguously in real time is crucial. Near infrared-I (NIRI) fluorescence-emitting targeting biomolecular constructs such as anticancer antibody-fluorophore conjugates, namely cetuximab-IRDye® 800CW (CTB-IRDye® 800CW), are FDA-approved for clinical trial usage in the fluorescence-guided resection of diseased tissue due to affording improved direct visualization of tumor tissue when compared to the use of either the unaided eye under standard white light illumination (WLI) surgical techniques or non-targeting fluorophores. Unfortunately, though helpful, CTB-IRDye® 800CW affords limited (i) identification of diseased tissue and (ii) tumor margin delineation, because the immunoconjugate generates suboptimal tumor-to-background ratios (TBRs) as a result of its spectral/photophysical profiles poorly aligning with the fixed optical windows of pre-/clinical setups. As such, CTB-IRDye® 800CW is more prone to affording incomplete resection compared to if TBRs were higher due to otherwise. To aid in accurately identifying deep-seated diseased tissue, photoacoustic (PA) tomography has been implemented alongside CTB-IRDye® 800CW to achieve PA signals that could result in higher TBRs. However, in clinical trial practice, using IRDye® 800CW for PA imaging also yields subpar TBRs due to it affording low PA signals. To overcome such limitations, we developed NIRDye 812, a structurally-modified topological equivalent of IRDye® 800CW, to confer it the capability to yield both higher TBRs and superior PA signal than that of the equivalent CTB-conjugate and fluorophore IRDye® 800CW itself, respectively. To do so, we substituted the oxygen atom at its meso-position with a sulfur atom. CTB-NIRDye 812 demonstrated a red-shifted absorption wavelength at 796 nm and a peak NIR-I fluorescence emission wavelength at 820 nm, which better dovetails with the fixed windows of preinstalled fixed emission filters within commercial pre-/clinical NIR-I fluorescence imaging instruments. Overall, CTB-NIRDye 812 provided a âˆ¼ 2-fold increase in TBRs compared to those of CTB-IRDye® 800CW in vivo. Also, NIRDye 812 displayed an ∼60% higher PA signal than that of IRDye® 800CW. Collectively, we achieved our goal of improving upon the spectral/photophysical and PA properties of IRDye® 800CW via introducing a subtle modification to its electronic core such that its CTB immunoconjugate could potentially allow for fast track or breakthrough designation by the FDA due to its near-identical structure displaying considerably improved efficacy.


Subject(s)
Fluorescent Dyes , Tomography, X-Ray Computed , Spectrum Analysis
4.
Chem Sci ; 13(44): 13201-13211, 2022 Nov 16.
Article in English | MEDLINE | ID: mdl-36425495

ABSTRACT

Fluorescence emission in the near-infrared-II (NIR-II) optical window affords reduced autofluorescence and light scattering, enabling deep-tissue visualization for both disease detection and surgical navigation. Small-molecule NIR-II dyes are preferable for clinical bioimaging applications, as the flexibility in their molecular synthesis allows for precise control of their optical and pharmacokinetic properties. Among the various types of dye, donor-acceptor-donor-based (D-A-D) dyes demonstrate exceptional photostability, whereas the frequently used PEGylation approach does not keep their intrinsic brightness enough in water environments due to their inherent effect of self-assembly. Here, we demonstrate that the commercially-available surfactants can serve as a dispersant to prevent molecular aggregation of PEGylated D-A-D dyes. Due to the favorable energetics for co-assembly between D-A-D dyes and surfactants, the formed surfactant-chaperoned dye strategy dramatically increases dye brightness. Accordingly, this effect provides remarkably improved performance for in vivo bioimaging applications. In parallel, we also investigate the D-A-D dye uptake and signal enhancement properties in the liver of murine models and demonstrate that the lumen-lining Kupffer cells can potentially disassemble PEGylated D-A-D aggregates such that their inherent brightness is restored. This phenomenon is similar to the surfactant-chaperoned dye strategy and our investigations provide a positive addition to better use of the current NIR-II fluorophores, especially for visualizing high-brightness required events.

5.
Nano Lett ; 22(23): 9596-9605, 2022 12 14.
Article in English | MEDLINE | ID: mdl-36394551

ABSTRACT

Clinical serology assays for detecting the antibodies of the virus are time-consuming, are less sensitive/selective, or rely on sophisticated detection instruments. Here, we develop a sandwiched plasmonic biosensor (SPB) for supersensitive thickness-sensing via utilizing the distance-dependent electromagnetic coupling in sandwiched plasmonic nanostructures. SPBs quantitatively amplify the thickness changes on the nanoscale range (sensitivity: ∼2% nm-1) into macroscopically visible signals, thereby enabling the rapid, label-free, and naked-eye detection of targeted biomolecular species (via the thickness change caused by immunobinding events). As a proof of concept, this assay affords a broad dynamic range (7 orders of magnitude) and a low LOD (∼0.3 pM), allowing for the extremely accurate SARS-CoV-2 antibody quantification (sensitivity/specificity: 100%/∼99%, with a portable optical fiber device). This strategy is suitable for high-throughput multiplexed detection and smartphone-based sensing at the point-of-care, which can be expanded for various sensing applications beyond the fields of viral infections and vaccination.


Subject(s)
Biosensing Techniques , COVID-19 , Humans , Surface Plasmon Resonance , Gold/chemistry , SARS-CoV-2 , COVID-19/diagnosis
6.
FEMS Microbiol Rev ; 45(6)2021 11 23.
Article in English | MEDLINE | ID: mdl-34160586

ABSTRACT

Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can lead to coronavirus disease 2019 (COVID-19). Virus-specific immunity controls infection, transmission and disease severity. With respect to disease severity, a spectrum of clinical outcomes occur associated with age, genetics, comorbidities and immune responses in an infected person. Dysfunctions in innate and adaptive immunity commonly follow viral infection. These are heralded by altered innate mononuclear phagocyte differentiation, activation, intracellular killing and adaptive memory, effector, and regulatory T cell responses. All of such affect viral clearance and the progression of end-organ disease. Failures to produce effective controlled antiviral immunity leads to life-threatening end-organ disease that is typified by the acute respiratory distress syndrome. The most effective means to contain SARS-CoV-2 infection is by vaccination. While an arsenal of immunomodulators were developed for control of viral infection and subsequent COVID-19 disease, further research is required to enable therapeutic implementation.


Subject(s)
COVID-19 , Adaptive Immunity , Humans , Immunity, Innate , SARS-CoV-2
7.
Front Mol Biosci ; 8: 670251, 2021.
Article in English | MEDLINE | ID: mdl-34026844

ABSTRACT

Thrombosis within the vasculature arises when pathological factors compromise normal hemostasis. On doing so, arterial thrombosis (AT) and venous thrombosis (VT) can lead to life-threatening cardio-cerebrovascular complications. Unfortunately, the therapeutic window following the onset of AT and VT is insufficient for effective treatment. As such, acute AT is the leading cause of heart attacks and constitutes ∼80% of stroke incidences, while acute VT can lead to fatal therapy complications. Early lesion detection, their accurate identification, and the subsequent appropriate treatment of thrombi can reduce the risk of thrombosis as well as its sequelae. As the success rate of therapy of fresh thrombi is higher than that of old thrombi, detection of the former and accurate identification of lesions as thrombi are of paramount importance. Magnetic resonance imaging, x-ray computed tomography (CT), and ultrasound (US) are the conventional non-invasive imaging modalities used for the detection and identification of AT and VT, but these modalities have the drawback of providing only image-delayed indirect visualization of only late stages of thrombi development. To overcome such limitations, near-infrared (NIR, ca. 700-1,700 nm) fluorescence (NIRF) imaging has been implemented due to its capability of providing non-invasive real-time direct visualization of biological structures and processes. Contrast agents designed for providing real-time direct or indirect visualization of thrombi using NIRF imaging primarily provide peak NIR-I fluorescence emission (ca. 700-1,000 nm), which affords limited tissue penetration depth and suboptimal spatiotemporal resolution. To facilitate the enhancement of the visualization of thrombosis via providing detection of smaller, fresh, and/or deep-seated thrombi in real time, the development of contrast agents with peak NIR-II fluorescence emission (ca. 1000-1,700 nm) has been recently underway. Currently, however, most contrast agents that provide peak NIR-II fluorescence emissions that are purportedly capable of providing direct visualization of thrombi or their resultant occlusions actually afford only the indirect visualization of such because they only provide for the (i) measuring of the surrounding vascular blood flow and/or (ii) simple tracing of the vasculature. These contrast agents do not target thrombi or occlusions. As such, this mini review summarizes the extremely limited number of targeting contrast agents with peak NIR-II fluorescence emission developed for non-invasive real-time direct visualization of thrombosis that have been recently reported.

8.
Adv Ther (Weinh) ; 4(5)2021 May.
Article in English | MEDLINE | ID: mdl-33997270

ABSTRACT

Thrombosis is an adverse physiological event wherein the resulting thrombus and thrombus-induced diseases collectively result in high morbidity and mortality rates. Currently, nano-medicines that incorporate fluorophores emitting in the near-infrared-I (NIR-I, 700-900 nm) spectral region into their systems have been adopted to afford thrombosis theranostics. However, several unsolved problems such as limited penetration depth and image quality severely impede further applications of such nano-medicine systems. Fortunately, the ability to incorporate fluorophores emitting in the NIR-II (1000-1700 nm) window into nano-medicine systems can unambiguously identify biological processes with high signal-to-noise, deep tissue penetration depth, and high image resolution. Considering the inherently favorable properties of NIR-II fluorophores, we believe such have enormous potential to quickly become incorporated into nano-medicine systems for thrombosis theranostics. In this review, we i) discuss the development of NIR fluorescence as an imaging modality and fluorescent agents; ii) comprehensively summarize the recent development of NIR-I fluorophore-based nano-medicine systems for thrombosis theranostics; iii) highlight the state-of-the-art NIR-II fluorophores that have been designed for the specific purpose of affording thrombotic diagnosis; iv) speculate on possible forward avenues for the use of NIR-II fluorophores towards thrombosis diagnosis and therapy; and v) discuss the potential for their clinical translation.

9.
Cancer Rep (Hoboken) ; 4(5): e1384, 2021 10.
Article in English | MEDLINE | ID: mdl-33811473

ABSTRACT

BACKGROUND: Tumor hypoxia is a characteristic of paramount importance due to low oxygenation levels in tissue negatively correlating with resistance to traditional therapies. The ability to noninvasively identify such could provide for personalized treatment(s) and enhance survival rates. Accordingly, we recently developed an NIR fluorescent hypoxia-sensitive smart probe (NO2 -Rosol) for identifying hypoxia via selectively imaging nitroreductase (NTR) activity, which could correlate to oxygen deprivation levels in cells, thereby serving as a proxy. We demonstrated proof of concept by subjecting a glioblastoma (GBM) cell line to extreme stress by evaluating such under radiobiological hypoxic (pO2 ≤ ~0.5%) conditions, which is a far cry from representative levels for hypoxia for brain glioma (pO2  = ~1.7%) which fluctuate little from physiological hypoxic (pO2  = 1.0-3.0%) conditions. AIM: We aimed to evaluate the robustness, suitability, and feasibility of NO2 -Rosol for imaging hypoxia in vitro and in vivo via assessing NTR activity in diverse GBM models under relevant oxygenation levels (pO2  = 2.0%) within physiological hypoxic conditions that mimic oxygenation levels in GBM tumor tissue in the brain. METHODS: We evaluated multiple GBM cell lines to determine their relative sensitivity to oxygenation levels via measuring carbonic anhydrase IX (CAIX) levels, which is a surrogate marker for indirectly identifying hypoxia by reporting on oxygen deprivation levels and upregulated NTR activity. We evaluated for hypoxia via measuring NTR activity when employing NO2 -Rosol in in vitro and tumor hypoxia imaging studies in vivo. RESULTS: The GBM39 cell line demonstrated the highest CAIX expression under hypoxic conditions representing that of GBM in the brain. NO2 -Rosol displayed an 8-fold fluorescence enhancement when evaluated in GBM39 cells (pO2  = 2.0%), thereby establishing its robustness and suitability for imaging hypoxia under relevant physiological conditions. We demonstrated the feasibility of NO2 -Rosol to afford tumor hypoxia imaging in vivo via it demonstrating a tumor-to-background of 5 upon (i) diffusion throughout, (ii) bioreductive activation by NTR activity in, and (iii) retention within, GBM39 tumor tissue. CONCLUSION: We established the robustness, suitability, and feasibility of NO2 -Rosol for imaging hypoxia under relevant oxygenation levels in vitro and in vivo via assessing NTR activity in GBM39 models.


Subject(s)
Fluorescence , Fluorescent Dyes/metabolism , Glioblastoma/pathology , Microscopy, Fluorescence/methods , Tumor Hypoxia , Animals , Apoptosis , Cell Proliferation , Female , Glioblastoma/diagnostic imaging , Glioblastoma/metabolism , Humans , Mice , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
J Neuroimmune Pharmacol ; 16(2): 270-288, 2021 06.
Article in English | MEDLINE | ID: mdl-33544324

ABSTRACT

Extracellular vesicles (EVs) are the common designation for ectosomes, microparticles and microvesicles serving dominant roles in intercellular communication. Both viable and dying cells release EVs to the extracellular environment for transfer of cell, immune and infectious materials. Defined morphologically as lipid bi-layered structures EVs show molecular, biochemical, distribution, and entry mechanisms similar to viruses within cells and tissues. In recent years their functional capacities have been harnessed to deliver biomolecules and drugs and immunological agents to specific cells and organs of interest or disease. Interest in EVs as putative vaccines or drug delivery vehicles are substantial. The vesicles have properties of receptors nanoassembly on their surface. EVs can interact with specific immunocytes that include antigen presenting cells (dendritic cells and other mononuclear phagocytes) to elicit immune responses or affect tissue and cellular homeostasis or disease. Due to potential advantages like biocompatibility, biodegradation and efficient immune activation, EVs have gained attraction for the development of treatment or a vaccine system against the severe acute respiratory syndrome coronavirus 2 (SARS CoV-2) infection. In this review efforts to use EVs to contain SARS CoV-2 and affect the current viral pandemic are discussed. An emphasis is made on mesenchymal stem cell derived EVs' as a vaccine candidate delivery system.


Subject(s)
COVID-19 Drug Treatment , Drug Delivery Systems/trends , Extracellular Vesicles , SARS-CoV-2/drug effects , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/metabolism , COVID-19/immunology , COVID-19/metabolism , Drug Delivery Systems/methods , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Humans , Immunity, Cellular/drug effects , Immunity, Cellular/physiology , Immunologic Factors/administration & dosage , Immunologic Factors/metabolism , SARS-CoV-2/immunology , SARS-CoV-2/metabolism
11.
Adv Drug Deliv Rev ; 171: 215-239, 2021 04.
Article in English | MEDLINE | ID: mdl-33428995

ABSTRACT

The SARS-CoV-2 global pandemic has seen rapid spread, disease morbidities and death associated with substantive social, economic and societal impacts. Treatments rely on re-purposed antivirals and immune modulatory agents focusing on attenuating the acute respiratory distress syndrome. No curative therapies exist. Vaccines remain the best hope for disease control and the principal global effort to end the pandemic. Herein, we summarize those developments with a focus on the role played by nanocarrier delivery.


Subject(s)
COVID-19 Vaccines/administration & dosage , COVID-19/prevention & control , Drug Carriers/administration & dosage , Nanocapsules/administration & dosage , SARS-CoV-2/drug effects , Animals , COVID-19/immunology , COVID-19 Vaccines/immunology , Drug Delivery Systems/methods , Drug Delivery Systems/trends , Humans , SARS-CoV-2/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology
12.
Sens Actuators B Chem ; 3062020 Mar 01.
Article in English | MEDLINE | ID: mdl-32265579

ABSTRACT

Hypoxia (pO2 ≤ ~1.5%) is an important characteristic of tumor microenvironments that directly correlates with resistance against first-line therapies and tumor proliferation/infiltration. The ability to accurately identify hypoxic tumor cells/tissue could afford tailored therapeutic regimens for personalized treatment, development of more-effective therapies, and discerning the mechanisms underlying disease progression. Fluorogenic constructs identifying aforesaid cells/tissue operate by targeting the bioreductive activity of primarily nitroreductases (NTRs), but collectively present photophysical and/or physicochemical shortcomings that could limit effectiveness. To overcome these limitations, we present the rational design, development, and evaluation of the first activatable ultracompact xanthene core-based molecular probe (NO 2 -Rosol) for selectively imaging NTR activity that affords an "OFF-ON" near-infrared (NIR) fluorescence response (> 700 nm) alongside a remarkable Stokes shift (> 150 nm) via NTR activity-facilitated modulation to its energetics whose resultant interplay discontinues an intramolecular d-PET fluorescence-quenching mechanism transpiring between directly-linked electronically-uncoupled π-systems comprising its components. DFT calculations guided selection of a suitable fluorogenic scaffold and nitroaromatic moiety candidate that when adjoined could (i) afford such photophysical response upon bioreduction by upregulated NTR activity in hypoxic tumor cells/tissue and (ii) employ a retention mechanism strategy that capitalizes on an inherent physical property of the NIR fluorogenic scaffold for achieving signal amplification. NO 2 -Rosol demonstrated 705 nm NIR fluorescence emission and 157 nm Stokes shift, selectivity for NTR over relevant bioanalytes, and a 28-/12-fold fluorescence enhancement in solution and between cells cultured under different oxic conditions, respectively. In establishing feasibility for NO 2 -Rosol to provide favorable contrast levels in solutio/vitro, we anticipate NO 2 -Rosol doing so in preclinical studies.

13.
Adv Mater ; 32(11): e1907365, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32022975

ABSTRACT

Tumor-lymph node (LN) metastasis is the dominant prognostic factor for tumor staging and therapeutic decision-making. However, concurrently visualizing metastasis and performing imaging-guided lymph node surgery is challenging. Here, a multiplexed-near-infrared-II (NIR-II) in vivo imaging system using nonoverlapping NIR-II probes with markedly suppressed photon scattering and zero-autofluorescence is reported, which enables visualization of the metastatic tumor and the tumor metastatic proximal LNs resection. A bright and tumor-seeking donor-acceptor-donor (D-A-D) dye, IR-FD, is screened for primary/metastatic tumor imaging in the NIR-IIa (1100-1300 nm) window. This optimized D-A-D dye exhibits greatly improved quantum yield of organic D-A-D fluorophores in aqueous solutions (≈6.0%) and good in vivo performance. Ultrabright PbS/CdS core/shell quantum dots (QDs) with dense polymer coating are used to visualize cancer-invaded sentinel LNs in the NIR-IIb (>1500 nm) window. Compared to clinically used indocyanine green, the QDs show superior brightness and photostability (no obvious bleaching even after continuous laser irradiation for 5 h); thus, only a picomolar dose is required for sentinel LNs detection. This combination of dual-NIR-II image-guided surgery can be performed under bright light, adding to its convenience and appeal in clinical use.


Subject(s)
Fluorescent Dyes/chemistry , Lymphatic Metastasis/diagnostic imaging , Optical Imaging/methods , Quantum Dots/chemistry , Sentinel Lymph Node/diagnostic imaging , Animals , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Cadmium Compounds/chemistry , Cell Line, Tumor , Female , Lead/chemistry , Lymphatic Metastasis/therapy , Mice , Polymers/chemistry , Selenium Compounds/chemistry , Sentinel Lymph Node/surgery , Spectroscopy, Near-Infrared/methods , Surgery, Computer-Assisted/methods
14.
RSC Adv ; 10(69): 42413-42422, 2020 Nov 22.
Article in English | MEDLINE | ID: mdl-33391732

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive form of primary brain tumor type and is associated with a high mortality rate borne out of such affording a survival rate of only 15 months. GBM aggressiveness is associated with the overexpression of epidermal growth factor receptor (EGFR) and its mutants. Targeting GBM with therapeutics is challenging because the blood-brain barrier (BBB) permits primarily select small-molecule entities across its semipermeable blockade. However, recent preclinical data suggest that large biomolecules, such as the anti-EGFR antibody therapeutic, cetuximab, could be capable of bypassing the BBB despite the relative enormity of its size. As such, we set forth to establish the feasibility of utilizing an EGFR-targeting near-infrared-I (NIR-I) fluorescent construct in the form of an immunoconjugate (cetuxmimab-IRDye800) to achieve visual differentiation between diseased brain tissue arising from a low-passage patient-derived GBM cell line (GBM39) and healthy brain tissue via utilizing orthotopic intracranial murine GBM39 tumor models for in vivo and ex vivo evaluation such that by doing so would establish proof of concept for ultimately facilitating its in vivo fluorescence-guided resection and ex vivo surgical back-table pathological confirmation in the clinic. As anticipated, we were not capable of distinguishing between malignant tumor tissue and healthy tissue in resected intact and slices of whole brain ex vivo under white-light illumination (WLI) due to both the diseased tissue and healthy tissue appearing virtually identical to the unaided eye. However, we readily observed over an average 6-fold enhancement in the fluorescence emission in the resected intact whole brain ex vivo when performing NIR-I fluorescence imaging (FLI) on the cohort of GBM39 tumor models that were administered the immunoconjugate compared to controls. In all, we laid the initial groundwork for establishing that NIR-I fluorescent immunoconjugates (theranostics) such as cetuximab-IRDye800 can bypass the BBB to visually afford GBM39 tumor tissue differentiation for its image-guided surgical removal.

15.
ACS Appl Bio Mater ; 3(12): 8658-8666, 2020 Dec 21.
Article in English | MEDLINE | ID: mdl-35019636

ABSTRACT

The primary treatment for malignant tumors remains to be resection. The strongest predictor of recurrence and postoperative prognosis is whether diseased tissue/cells remain(s) at the surgical margin. Cancer surgery entails surgeons having the capability to visually distinguish between subtle shades of color in attempts of differentiating between diseased tissue and healthy tissue under standard white-light illumination, as such tissue states appear identical at the meso-/macroscopic level. Accordingly, enhancing the capability of surgeons to do so such that they can accurately delineate the tumor margin is of paramount importance. Fluorescence-guided surgery facilitates in enhancing such capability by color-coding the surgical field with overlaid contrasting pseudo-colors from real-time intraoperative fluorescence emission via utilizing fluorescent constructs in tandem. Constructs undergoing clinical trials or that are FDA-approved provide peak fluorescence emission in the visible (405 - 700 nm) or near-infrared-I (NIR-I) spectral region (700-900 nm), whereby differentiation between tissue states progressively improves in sync with using constructs that emit longer wavelengths of light. Here, we repurpose the usage of such fluorescent constructs by establishing feasibility of a tumor-targeting immunoconjugate (cetuximab-IRDye800) having peak fluorescence emission at the NIR-I spectral region to provide improved tumor margin delineation by affording higher tumor-to-background ratios (TBRs) when measuring its off-peak fluorescence emission at the near-infrared-II (NIR-II) spectral region (1000-1700 nm) in in vivo applications. We prepared murine tumor models, administered such immunoconjugate, and imaged such models pre-/post-administration via utilizing imaging systems that separately afforded acquisition of fluorescence emission in the NIR-I or NIR-II spectral region. On doing so, we determined in vivo TBRs, ex vivo TBRs with/-out skin, and ex vivo biodistribution, all via measuring the fluorescence emission of the immunoconjugate at tumor site(s) at both spectral regions. Collectively, we established feasibility of using the immunoconjugate to afford improved tumor margin delineation by providing 2-fold higher TBRs via utilizing the NIR-II spectral region to capture off-peak fluorescence emission from a fluorescent construct having NIR-I peak fluorescence emission.

16.
Sci Adv ; 5(9): eaaw0672, 2019 09.
Article in English | MEDLINE | ID: mdl-31548981

ABSTRACT

NIR-II fluorescence imaging greatly reduces scattering coefficients for nearly all tissue types at long wavelengths, benefiting deep tissue imaging. However, most of the NIR-II fluorophores suffer from low quantum yields and/or short circulation time that limit the quality of NIR-II imaging. Here, we engineered a supramolecular assembly of protein complex with lodged cyanine dyes to produce a brilliant NIR-II fluorophore, providing a NIR-II quantum yield of 21.2% with prolonged circulation time. Computational modeling revealed the mechanism for fluorescence enhancement and identified key parameters governing albumin complex for NIR-II fluorophores. Our complex afforded high-resolution microvessel imaging, with a 3-hour imaging window compared to 2 min for free dye alone. Furthermore, the complexation strategy was applied to an antibody-derived assembly, offering high-contrast tumor imaging without affecting the targeting ability of the antibody. This study provides a facile strategy for producing high-performance NIR-II fluorophores by chaperoning cyanine dyes with functional proteins.


Subject(s)
Contrast Media , Fluorescent Dyes , Neoplasms, Experimental , Optical Imaging , Serum Albumin, Bovine , Animals , Cattle , Contrast Media/chemistry , Contrast Media/pharmacokinetics , Contrast Media/pharmacology , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacokinetics , Fluorescent Dyes/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/diagnostic imaging , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/pharmacokinetics , Serum Albumin, Bovine/pharmacology
17.
Anal Chem ; 91(4): 3110-3117, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30669835

ABSTRACT

An optical molecular imaging contrast agent that is tailored toward lymphatic mapping techniques implementing near-infrared (NIR) fluorescence image-guided navigation in the planning and surgical treatment of cancers would significantly aid in enabling the real-time visualization of the potential metastatic tumor-draining lymph node(s) for their needed surgical biopsy and/or removal, thereby ensuring unmissed disease to prevent recurrence and improve patient survival rates. Here, the development of the first NIR fluorescent rosol dye (THQ-Rosol) tailored to overcome the limitations arising from the suboptimal properties of the generic molecular fluorescent dyes commonly used for such applications is described. In developing THQ-Rosol, we prepared a progressive series of torsionally restrictive N-substituted non-NIR fluorescent rosol dyes based on density functional theory (DFT) calculations, wherein we discerned high correlations amongst their calculated energetics, modeled N-C3' torsion angles, and evaluated properties. We leveraged these strong relationships to rationally design THQ-Rosol, wherein DFT calculations inspired an innovative approach and synthetic strategy to afford an uncharged xanthene core-based scaffold/molecular platform with an aptly elevated p Ka value alongside NIR fluorescence emission (ca.700-900 nm). THQ-Rosol exhibited 710 nm NIR fluorescence emission, a 160 nm Stokes shift, robust photostability, and an aptly elevated p Ka value (5.85) for affording pH-insensitivity and optimal contrast upon designed use. We demonstrated the efficacy of THQ-Rosol for lymphatic mapping with in vitro and in vivo studies, wherein it revealed timely tumor drainage and afforded definitive lymph node visualization upon its administration and accumulation. THQ-Rosol serves as a proof-of-concept for the effective tailoring of an uncharged xanthene core-based scaffold/molecular platform toward a specific imaging application using rational design.


Subject(s)
Diamines/chemistry , Fluorescent Dyes/chemistry , Lymph Nodes/diagnostic imaging , Lymphatic Metastasis/diagnostic imaging , Optical Imaging , Diamines/chemical synthesis , Fluorescence , Fluorescent Dyes/chemical synthesis , Humans , Infrared Rays , Molecular Structure
18.
ACS Chem Neurosci ; 7(1): 21-5, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26521705

ABSTRACT

A molecular imaging tool that provides for the direct visualization of serotonin would significantly aid in the investigation of neuropsychiatric disorders that are attributed to its neuronal dysregulation. Here, the design, synthesis, and evaluation of NeuroSensor 715 (NS715) is presented. NS715 is the first molecular sensor that exhibits a turn-on near-infrared fluorescence response toward serotonin. Density functional theory calculations facilitated the design of a fluorophore based on a coumarin-3-aldehyde scaffold that derives from an electron-rich 1,2,3,4-tetrahydroquinoxaline framework, which provides appropriate energetics to prevent the hydroxyindole moiety of serotonin from quenching its fluorescence emission. Spectroscopic studies revealed that NS715 produces an 8-fold fluorescence enhancement toward serotonin with an emission maximum at 715 nm. Accompanying binding studies indicated NS715 displays a 19-fold selective affinity for serotonin and a modest affinity for catecholamines over other primary-amine neurotransmitters. The utility of NS715 toward neuroimaging applications was validated by selectively labeling and directly imaging norepinephrine within secretory vesicles using live chromaffin cells, which serve as a model system for specialized neurons that synthesize, package, and release only a single, unique type of neurotransmitter. In addition, NS715 effectively differentiated between cell populations that express distinct neurotransmitter phenotypes.


Subject(s)
Chromaffin Cells/metabolism , Molecular Imaging , Serotonin/analysis , Animals , Chromaffin Cells/chemistry , Chromaffin Cells/cytology , Dose-Response Relationship, Drug , Epinephrine/metabolism , Fluorescent Dyes/pharmacokinetics , Glutamic Acid/metabolism , Secretory Vesicles/drug effects , Secretory Vesicles/metabolism , Spectrometry, Fluorescence , Sulfates/pharmacology
19.
Chemistry ; 21(32): 11446-51, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26119241

ABSTRACT

Tunable dual-analyte fluorescent molecular logic gates (ExoSensors) were designed for the purpose of imaging select vesicular primary-amine neurotransmitters that are released from secretory vesicles upon exocytosis. ExoSensors are based on the coumarin-3-aldehyde scaffold and rely on both neurotransmitter binding and the change in environmental pH associated with exocytosis to afford a unique turn-on fluorescence output. A pH-functionality was directly integrated into the fluorophore π-system of the scaffold, thereby allowing for an enhanced fluorescence output upon the release of labeled neurotransmitters. By altering the pH-sensitive unit with various electron-donating and -withdrawing sulfonamide substituents, we identified a correlation between the pKa of the pH-sensitive group and the fluorescence output from the activated fluorophore. In doing so, we achieved a twelvefold fluorescence enhancement upon evaluating the ExoSensors under conditions that mimic exocytosis. ExoSensors are aptly suited to serve as molecular imaging tools that allow for the direct visualization of only the neurotransmitters that are released from secretory vesicles upon exocytosis.


Subject(s)
Coumarins/chemistry , Exocytosis/drug effects , Fluorescent Dyes/chemistry , Neurons/chemistry , Neurotransmitter Agents/chemistry , Computers, Molecular , Coumarins/metabolism , Neurons/metabolism , Neurotransmitter Agents/metabolism , Optical Imaging
20.
Chemistry ; 20(52): 17488-99, 2014 Dec 22.
Article in English | MEDLINE | ID: mdl-25346467

ABSTRACT

NeuroSensor 521 (NS521) is a fluorescent sensor for primary-amine neurotransmitters based on a platform that consists of an aryl moiety appended to position C4 of the coumarin-3-aldehyde scaffold. We demonstrate that sensors based on this platform behave as a directly linked donor-acceptor system that operates through an intramolecular acceptor-excited photoinduced electron transfer (a-PET) mechanism. To evaluate the PET process, a series of benzene- and thiophene-substituted derivatives were prepared and the photophysical properties, binding affinities, and fluorescence responses toward glutamate, norepinephrine, and dopamine were determined. The calculated energy of the highest occupied molecular orbital (EHOMO ) of the pendant aryl substituents, along with oxidation and reduction potential values derived from the calculated molecular orbital energy values of the platform components, allowed for calculation of the fluorescence properties of the benzene sensor series. Interestingly, the thiophene derivatives did not fit the typical PET model, highlighting the limitations of the method. A new sensor, NeuroSensor 539, displayed enhanced photophysical properties aptly suited for biological imaging. NeuroSensor 539 was validated by selectively labeling and imaging norepinephrine in secretory vesicles of live chromaffin cells.


Subject(s)
Aldehydes/chemistry , Chromaffin Cells/chemistry , Coloring Agents/chemistry , Coumarins/chemistry , Fluorescent Dyes/chemistry , Neurotransmitter Agents/chemistry , Norepinephrine/chemistry , Electron Transport , Oxidation-Reduction , Positron-Emission Tomography , Quantum Theory , Spectrometry, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL
...