Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Mol Pharm ; 21(1): 137-142, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37989273

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) remains one of the most dismal diagnoses that a patient can receive. PDAC is extremely difficult to treat, as drug delivery is challenging in part due to the lack of vascularization, high stromal content, and high collagen content of these tumors. We have previously demonstrated that attaching drugs to the cobalamin scaffold provides selectivity for tumors over benign cells due to a high vitamin demand in these rapidly growing cells and an overexpression of transcobalamin receptors in a variety of cancer types. Importantly, we have shown the ability to deliver cobalamin derivatives to orthotopic pancreas tumors. Tyrosine kinase inhibitors have shown promise in treating PDAC as well as other cancer types. However, some of these inhibitors suffer from drug resistance, and as such, their success has been diminished. With this in mind, we synthesized the tyrosine kinase inhibitors erlotinib (EGFR) and dasatinib (Src) that are attached to this cobalamin platform. Both of these cobalamin-drug conjugates cause visible light-induced apoptosis, and the cobalamin-erlotinib conjugate (2) causes X-ray-induced apoptosis in MIA PaCa-2 cells. Both visible light and X-rays provide spatial control of drug release; however, utilizing X-ray irradiation offers the advantage of deeper tissue penetration. Therefore, we explored the utilization of 2 as a synergistic therapy with radiation in athymic nude mice implanted with MIA PaCa-2 tumors. We discovered that the addition of 2 caused an enhanced reduction in tumor margins in comparison with radiation therapy alone. In addition, treatment with 2 in the absence of radiation caused no significant reduction in tumor size in comparison with the controls. The cobalamin technology presented here allows for the spatial release of drugs in conjunction with external beam radiation therapy, potentially allowing for more effective treatment of deep-seated tumors with less systemic side effects.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Mice , Animals , Humans , Erlotinib Hydrochloride/pharmacology , Erlotinib Hydrochloride/therapeutic use , Vitamin B 12/therapeutic use , Mice, Nude , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/radiotherapy , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/radiotherapy , Pancreatic Neoplasms/pathology , Cell Line, Tumor
2.
Opt Lett ; 45(22): 6130-6133, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-33186932

ABSTRACT

Cherenkov light induced from megavolt (MV) X-rays during external beam radiotherapy serves as an internal light source to excite phosphors or fluorophores within biological tissues for molecular imaging. The broad spectrum of Cherenkov light leads to significant spectral overlap with any luminescence emission and, to overcome this problem, a single pixel hyperspectral imaging methodology was demonstrated here by coupling the detection with light sheet scanning and filtered back projection reconstruction of hyperspectral images. Thin scanned sheets of MV X-rays produce Cherenkov light to illuminate the planes deep within the tissue-simulating media. A fluorescence probe was excited by Cherenkov light, and a complete hyperspectral sinogram of the data was obtained through translation and rotation of the beam. Hyperspectral 2D images finally were reconstructed. Through this approach of spectral unmixing, it was possible to resolve hyperspectral images of both the Cherenkov and resulting fluorescence intensity from molecular sensors.


Subject(s)
Optical Imaging/instrumentation , Particle Accelerators , Image Processing, Computer-Assisted , Surface Properties , X-Rays
3.
Opt Lett ; 45(3): 664, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-32004278

ABSTRACT

This publisher's note contains corrections to Opt. Lett.45, 284 (2020)OPLEDP0146-959210.1364/OL.45.000284.

4.
J Surg Oncol ; 122(8): 1711-1720, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32885452

ABSTRACT

BACKGROUND: Current practices for fluorescence-guided cancer surgery utilize a single fluorescent agent, but homogeneous distribution throughout the tumor is difficult to achieve. We hypothesize that administering a perfusion and a molecular-targeted agent at their optimal administration-to-imaging time will improve whole-tumor contrast. EXPERIMENTAL DESIGN: Mice bearing subcutaneous xenograft human synovial sarcomas were administered indocyanine green (ICG) (3 mg/kg) or ABY-029 (48.7 µg/kg)-an epidermal growth factor receptor-targeted Affibody molecule-alone or in combination. Fluorescence contrast and signal distribution were compared between treatment groups. Two commercial fluorescence imaging systems were tested for simultaneous imaging of ICG and ABY-029. RESULTS: ABY-029 has a moderate positive correlation with viable tumor (ρ = 0.2 ± 0.4), while ICG demonstrated a strong negative correlation (ρ = -0.6 ± 0.1). The contrast-to-variance ratio was highest in the ABY-029 +ICG (2.5 ± 0.8), compared to animals that received ABY-029 (2.3 ± 0.8) or ICG (2.0 ± 0.5) alone. Moreover, the combination of ABY-029 + ICG minimizes the correlation between viable tumor and fluorescence intensity (ρ = -0.1 ± 0.2) indicating the fluorescence signal distribution is more homogeneous throughout the tumor milieu. CONCLUSION: Dual-agent imaging utilizing a single channel in a commercial fluorescence-guided imaging system tailored for IRDye 800CW is a promising method to increase tumor contrast in a clinical setting.


Subject(s)
Fluorescence , Fluorescent Dyes/metabolism , Molecular Imaging/methods , Optical Imaging/methods , Recombinant Fusion Proteins/metabolism , Sarcoma/pathology , Animals , Cell Proliferation , Humans , Indocyanine Green , Mice , Sarcoma/diagnostic imaging , Sarcoma/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Opt Lett ; 44(7): 1552-1555, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30933088

ABSTRACT

Cherenkov-excited luminescence scanned imaging (CELSI) is achieved with a clinical linear accelerator during external beam radiotherapy to map out molecular luminescence intensity or lifetime in tissue. In order to realize a deeper imaging depth with a reasonable spatial resolution in CELSI, we optimized the original scanning procedure to complete this in a way similar to x-ray computed tomography and with image reconstruction from maximum-likelihood expectation maximization and multi-pinhole irradiation for parallelization. Resolution phantom studies showed that a 0.3 mm diameter capillary tube containing 0.01 nM luminescent nanospheres could be recognized at a depth of 21 mm into tissue-like media. Small animal imaging with a 1 mm diameter cylindrical target demonstrated that fast 3D data acquisition can be achieved by this multi-pinhole collimator approach to image high-resolution luminescence through a whole animal.


Subject(s)
Image Processing, Computer-Assisted/methods , Luminescence , Tomography/methods , Animals , Female , Mice , Phantoms, Imaging
6.
J Surg Oncol ; 119(8): 1077-1086, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30950072

ABSTRACT

BACKGROUND AND OBJECTIVES: Fluorescence-guided surgery using epidermal growth factor receptor (EGFR) targeting has been performed successfully in clinical trials using a variety of fluorescent agents. We investigate ABY-029 (anti-EGFR Affibody® molecule labeled with IRDye 800CW) compared with a small-molecule perfusion agent, IRDye 700DX carboxylate, in a panel of soft-tissue sarcomas with varying levels of EGFR expression and vascularization. METHODS: Five xenograft soft-tissue sarcoma cell lines were implanted into immunosuppressed mice. ABY-029 and IRDye 700DX were each administered at 4.98 µM. Fluorescence from in vivo and ex vivo (fresh and formalin-fixed) fixed tissues were compared. The performance of three fluorescence imaging systems was assessed for ex vivo tissues. RESULTS: ABY-029 is retained longer within tumor tissue and achieves higher tumor-to-background ratios both in vivo and ex vivo than IRDye 700DX. ABY-029 fluorescence is less susceptible to formalin fixation than IRDye 700DX, but both agents have disproportional signal loss in a variety of tissues. The Pearl Impulse provides the highest contrast-to-noise ratio, but all systems have individual advantages. CONCLUSIONS: ABY-029 demonstrates promise to assist in wide local excision of soft-tissue sarcomas. Further clinical evaluation of in situ or freshly excised ex vivo tissues using fluorescence imaging systems is warranted.


Subject(s)
ErbB Receptors/analysis , Molecular Probes , Recombinant Fusion Proteins , Sarcoma/diagnostic imaging , Sarcoma/surgery , Animals , Cell Line, Tumor , ErbB Receptors/biosynthesis , Female , Humans , Male , Mice , Optical Imaging/methods , Sarcoma/enzymology , Surgery, Computer-Assisted/methods , Xenograft Model Antitumor Assays
7.
Cancer Control ; 25(1): 1073274817752332, 2018.
Article in English | MEDLINE | ID: mdl-29334791

ABSTRACT

The excision of tumors by wide local excision is challenging because the mass must be removed entirely without ever viewing it directly. Positive margin rates in sarcoma resection remain in the range of 20% to 35% and are associated with increased recurrence and decreased survival. Fluorescence-guided surgery (FGS) may improve surgical accuracy and has been utilized in other surgical specialties. ABY-029, an anti-epidermal growth factor receptor Affibody molecule covalently bound to the near-infrared fluorophore IRDye 800CW, is an excellent candidate for future FGS applications in sarcoma resection; however, conventional methods with direct surface tumor visualization are not immediately applicable. A novel technique involving imaging through a margin of normal tissue is needed. We review the past and present applications of FGS and present a novel concept of indirect FGS for visualizing tumor through a margin of normal tissue and aiding in excising the entire lesion as a single, complete mass with tumor-free margins.


Subject(s)
Neoplasms/surgery , Surgery, Computer-Assisted/methods , Fluorescence , Humans
8.
J Biomech Eng ; 139(6)2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28388715

ABSTRACT

Elevated total tissue pressure (TTP) in pancreatic adenocarcinoma is often associated with stress applied by cellular proliferation and hydrated hyaluronic acid osmotic swelling; however, the causal roles of collagen in total tissue pressure have yet to be clearly measured. This study illustrates one direct correlation between total tissue pressure and increased deposition of collagen within the tissue matrix. This observation comes from a new modification to a conventional piezoelectric pressure catheter, used to independently separate and quantify total tissue pressure, solid stress (SS), and interstitial fluid pressure (IFP) within the same tumor location, thereby clarifying the relationship between these parameters. Additionally, total tissue pressure shows a direct correlation with verteporfin uptake, demonstrating the impediment of systemically delivered molecules with increased tissue hypertension.


Subject(s)
Collagen/metabolism , Extracellular Fluid/metabolism , Pancreatic Neoplasms/pathology , Pressure , Stress, Mechanical , Animals , Biological Transport , Cell Line, Tumor , Cell Transformation, Neoplastic , Compressive Strength , Female , Humans , Pancreatic Neoplasms/metabolism , Porphyrins/metabolism , Rats , Verteporfin , Pancreatic Neoplasms
9.
Proc Natl Acad Sci U S A ; 110(22): 9025-30, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23671066

ABSTRACT

The up-regulation of cell surface receptors has become a central focus in personalized cancer treatment; however, because of the complex nature of contrast agent pharmacokinetics in tumor tissue, methods to quantify receptor binding in vivo remain elusive. Here, we present a dual-tracer optical technique for noninvasive estimation of specific receptor binding in cancer. A multispectral MRI-coupled fluorescence molecular tomography system was used to image the uptake kinetics of two fluorescent tracers injected simultaneously, one tracer targeted to the receptor of interest and the other tracer a nontargeted reference. These dynamic tracer data were then fit to a dual-tracer compartmental model to estimate the density of receptors available for binding in the tissue. Applying this approach to mice with deep-seated gliomas that overexpress the EGF receptor produced an estimate of available receptor density of 2.3 ± 0.5 nM (n = 5), consistent with values estimated in comparative invasive imaging and ex vivo studies.


Subject(s)
Fluorescence , Glioma/metabolism , Magnetic Resonance Imaging/methods , Molecular Imaging/methods , Receptors, Cell Surface/metabolism , Tomography, Optical/methods , Animals , ErbB Receptors/metabolism , Kinetics , Mice , Protein Binding
10.
Opt Lett ; 40(5): 827-30, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25723443

ABSTRACT

Ionizing radiation is commonly delivered by medical linear accelerators (LINAC) in the form of shaped beams, and it is able to induce Cherenkov emission in tissue. In fluorescence-based microscopy excitation from scanned spots, lines, or sheets can be used for fast high-resolution imaging. Here we introduce Cherenkov-excited luminescence scanned imaging (CELSI) as a new imaging methodology utilizing 2-dimensional (∼5-mm-thick) sheets of LINAC radiation to produce Cherenkov photons, which in turn excite luminescence of probes distributed in biological tissues. Imaging experiments were performed by scanning these excitation sheets in three orthogonal directions while recording Cherenkov-excited luminescence. Tissue phantom studies have shown that single luminescent inclusions ∼1 mm in diameter can be imaged within 20-mm-thick tissue-like media with minimal loss of spatial resolution. Using a phosphorescent probe for oxygen, PtG4 with the CELSI methodology, an image of partial pressure of oxygen (pO2) was imaged in a rat lymph node, quantitatively restoring pO2 values in differently oxygenated tissues.


Subject(s)
Luminescence , Optical Imaging/methods , Animals , Optical Imaging/instrumentation , Particle Accelerators , Phantoms, Imaging , Photons , Rats
11.
Opt Lett ; 38(23): 5184-7, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24281541

ABSTRACT

Assessing tumor margin status during surgery is critical to ensure complete resection of cancer tissue; however, current approaches are ineffective and often result in repeat surgery. We present an optical imaging approach for margin assessment using topical application of two fluorescent stains, one targeted to a tumor biomarker and the other a nontargeted reference, to freshly excised specimens. Computing a normalized difference image from fluorescence images of the targeted and untargeted stains suppresses the confounding effects of nonspecific uptake. Applying this approach in excised breast tumor models produced promising tumor-to-normal tissue contrasts that were significantly higher than single-targeted-stain imaging.


Subject(s)
Fluorescent Dyes/administration & dosage , Fluorescent Dyes/metabolism , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/surgery , Optical Imaging/methods , Adipose Tissue/pathology , Administration, Topical , Animals , Intraoperative Period , Mammary Neoplasms, Experimental/metabolism , Mice , Time Factors
12.
Mol Imaging Biol ; 25(1): 110-121, 2023 02.
Article in English | MEDLINE | ID: mdl-34651290

ABSTRACT

PURPOSE: The goal of fluorescence-guided surgery (FGS) in oncology is to improve the surgical therapeutic index by enhancing contrast between cancerous and healthy tissues. However, optimal discrimination between these tissues is complicated by the nonspecific uptake and retention of molecular targeted agents and the variance of fluorescence signal. Paired-agent imaging (PAI) employs co-administration of an untargeted imaging agent with a molecular targeted agent, providing a normalization factor to minimize nonspecific and varied signals. The resulting measured binding potential is quantitative and equivalent to in vivo immunohistochemistry of the target protein. This study demonstrates that PAI improves the accuracy of tumor-to-healthy tissue discrimination compared to single-agent imaging for in vivo FGS. PROCEDURES: PAI using a fluorescent anti-epidermal growth factor receptor (EGFR) affibody molecule (ABY-029, eIND 122,681) with untargeted IRDye 700DX carboxylate was compared to ABY-029 alone in an oral squamous cell carcinoma xenograft mouse model at 3 h after dye administration (n = 30). RESULTS: PAI significantly enhanced tumor discrimination, as compared to ABY-029 alone in low EGFR-expressing tumors and highly heterogeneous populations including multiple cell lines with varying expression (diagnostic accuracy: 0.908 vs. 0.854 and 0.908 vs. 0.822; and ROC curve AUC: 0.963 vs. 0.909 and 0.957 vs. 0.909, respectively) indicating a potential for universal FGS image thresholds to determine surgical margins. In addition, PAI achieved significantly higher diagnostic ability than ABY-029 alone 0.25-5-h post injection and exhibited a stronger correlation to EGFR expression heterogeneity. CONCLUSION: The quantitative receptor delineation of PAI promises to improve the surgical therapeutic index of cancer resection in a clinically relevant timeline.


Subject(s)
Carcinoma, Squamous Cell , Mouth Neoplasms , Surgery, Computer-Assisted , Humans , Mice , Animals , Mouth Neoplasms/diagnostic imaging , Mouth Neoplasms/surgery , ErbB Receptors/metabolism , Surgery, Computer-Assisted/methods , Optical Imaging/methods , Cell Line, Tumor
13.
Front Med Technol ; 5: 1009638, 2023.
Article in English | MEDLINE | ID: mdl-36875185

ABSTRACT

Background: Fluorescence molecular imaging using ABY-029, an epidermal growth factor receptor (EGFR)-targeted, synthetic Affibody peptide labeled with a near-infrared fluorophore, is under investigation for surgical guidance during head and neck squamous cell carcinoma (HNSCC) resection. However, tumor-to-normal tissue contrast is confounded by intrinsic physiological limitations of heterogeneous EGFR expression and non-specific agent uptake. Objective: In this preliminary study, radiomic analysis was applied to optical ABY-029 fluorescence image data for HNSCC tissue classification through an approach termed "optomics." Optomics was employed to improve tumor identification by leveraging textural pattern differences in EGFR expression conveyed by fluorescence. The study objective was to compare the performance of conventional fluorescence intensity thresholding and optomics for binary classification of malignant vs. non-malignant HNSCC tissues. Materials and Methods: Fluorescence image data collected through a Phase 0 clinical trial of ABY-029 involved a total of 20,073 sub-image patches (size of 1.8 × 1.8 mm2) extracted from 24 bread-loafed slices of HNSCC surgical resections originating from 12 patients who were stratified into three dose groups (30, 90, and 171 nanomoles). Each dose group was randomly partitioned on the specimen-level 75%/25% into training/testing sets, then all training and testing sets were aggregated. A total of 1,472 standardized radiomic features were extracted from each patch and evaluated by minimum redundancy maximum relevance feature selection, and 25 top-ranked features were used to train a support vector machine (SVM) classifier. Predictive performance of the SVM classifier was compared to fluorescence intensity thresholding for classifying testing set image patches with histologically confirmed malignancy status. Results: Optomics provided consistent improvement in prediction accuracy and false positive rate (FPR) and similar false negative rate (FNR) on all testing set slices, irrespective of dose, compared to fluorescence intensity thresholding (mean accuracies of 89% vs. 81%, P = 0.0072; mean FPRs of 12% vs. 21%, P = 0.0035; and mean FNRs of 13% vs. 17%, P = 0.35). Conclusions: Optomics outperformed conventional fluorescence intensity thresholding for tumor identification using sub-image patches as the unit of analysis. Optomics mitigate diagnostic uncertainties introduced through physiological variability, imaging agent dose, and inter-specimen biases of fluorescence molecular imaging by probing textural image information. This preliminary study provides a proof-of-concept that applying radiomics to fluorescence molecular imaging data offers a promising image analysis technique for cancer detection in fluorescence-guided surgery.

14.
J Cell Mol Med ; 16(11): 2690-700, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22568538

ABSTRACT

PV1 is an endothelial-specific protein with structural roles in the formation of diaphragms in endothelial cells of normal vessels. PV1 is also highly expressed on endothelial cells of many solid tumours. On the basis of in vitro data, PV1 is thought to actively participate in angiogenesis. To test whether or not PV1 has a function in tumour angiogenesis and in tumour growth in vivo, we have treated pancreatic tumour-bearing mice by single-dose intratumoural delivery of lentiviruses encoding for two different shRNAs targeting murine PV1. We find that PV1 down-regulation by shRNAs inhibits the growth of established tumours derived from two different human pancreatic adenocarcinoma cell lines (AsPC-1 and BxPC-3). The effect observed is because of down-regulation of PV1 in the tumour endothelial cells of host origin, PV1 being specifically expressed in tumour vascular endothelial cells and not in cancer or other stromal cells. There are no differences in vascular density of tumours treated or not with PV1 shRNA, and gain and loss of function of PV1 in endothelial cells does not modify either their proliferation or migration, suggesting that tumour angiogenesis is not impaired. Together, our data argue that down-regulation of PV1 in tumour endothelial cells results in the inhibition of tumour growth via a mechanism different from inhibiting angiogenesis.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Carrier Proteins/genetics , Membrane Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Adenocarcinoma/blood supply , Animals , Base Sequence , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Down-Regulation , Drug Screening Assays, Antitumor , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Lentivirus/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mice, Nude , Molecular Sequence Data , Neovascularization, Pathologic/genetics , Pancreatic Neoplasms/blood supply , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Stromal Cells/metabolism , Stromal Cells/pathology
15.
Gastroenterology ; 141(3): 1091-101, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21699781

ABSTRACT

BACKGROUND & AIMS: Rb1 encodes a cell-cycle regulator that is functionally disrupted in most human cancers. Pancreatic ductal adenocarcinomas (PDACs) have a high frequency of mutations in KRAS and INK4A/CDKN2A that might allow cells to bypass the regulatory actions of retinoblastoma (RB). To determine the role of loss of RB function in PDAC progression, we investigated the effects of Rb disruption during pancreatic malignant transformation initiated by oncogenic Kras. METHODS: We generated mice with pancreas-specific disruption of Rb, in the absence or presence of oncogenic Kras, to examine the role of RB in pancreatic carcinogenesis. RESULTS: In the presence of oncogenic Kras, loss of Rb from the pancreatic epithelium accelerated formation of pancreatic intraepithelial neoplasia (PanIN), increased the frequency of cystic neoplasms, and promoted rapid progression toward PDAC. Early stage cancers were characterized by acute pancreatic inflammation, associated with up-regulation of proinflammatory cytokines within the pancreas. Despite the presence of markers associated with oncogene-induced senescence, low-grade PanIN were highly proliferative and expressed high levels of p53. Pancreatic cancer cell lines derived from these mice expressed high levels of cytokines, and transcriptional activity of p53 was impaired. CONCLUSIONS: Rb encodes a tumor suppressor that attenuates progression of oncogenic Kras-induced carcinogenesis in the pancreas by mediating the senescence response and promoting activity of the tumor suppressor p53.


Subject(s)
Carcinoma in Situ/physiopathology , Cellular Senescence/physiology , Gene Deletion , Pancreatic Neoplasms/physiopathology , Precancerous Conditions/physiopathology , Proto-Oncogene Proteins p21(ras)/physiology , Retinoblastoma Protein/genetics , Retinoblastoma Protein/physiology , Adenocarcinoma/pathology , Adenocarcinoma/physiopathology , Animals , Carcinoma in Situ/pathology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/physiopathology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Cytokines/physiology , Disease Models, Animal , Disease Progression , Mice , Mice, Mutant Strains , Pancreatic Neoplasms/pathology , Precancerous Conditions/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Suppressor Protein p53/physiology
16.
J Biomed Opt ; 26(1)2021 01.
Article in English | MEDLINE | ID: mdl-33423407

ABSTRACT

SIGNIFICANCE: The study has confirmed the feasibility of using ultraviolet (UV) excitation to visualize and quantify desmoplasia in fresh tumor tissue of pancreatic adenocarcinoma (PDAC) in an orthotopic xenograft mouse model, which provides a useful imaging platform to evaluate acute therapeutic responses. AIM: Stromal network of collagen prominent in PDAC tumors is examined by imaging fresh tissue samples stained with histological dyes. Fluorescence signals are color-transferred to mimic Masson's trichrome staining. APPROACH: Murine tumor samples were stained with Hoechst, eosin, and rhodamine B and excited at 275-nm. Fluorescence signals in the visible spectrum were captured by a CMOS color camera with high contrast and resolution at whole-tumor slice field of view. RESULTS: Fluorescence imaging using UV excitation is capable of visualizing collagen deposition in PDAC tumors. Both fluorescence and histology data showed collagen content of up to 30%. The collagen modulation effect due to photodynamic priming treatment was observed showing 13% of collagen reduction. Necrosis area is visible and perfusion imaging using Texas Red dextran is feasible. CONCLUSIONS: The study demonstrates collagen visualization in fresh PDAC tumor samples using UV excitation. This imaging platform also provides quantitative stromal information from fiber analysis and visibility of necrosis and perfusion, suitable for therapeutic response assessment of photodynamic therapy.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , Photochemotherapy , Animals , Collagen , Mice , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/drug therapy
17.
J Biophotonics ; 14(11): e202100088, 2021 11.
Article in English | MEDLINE | ID: mdl-34323374

ABSTRACT

We report a high light-throughput spectroscopic dosimeter system that is able to noninvasively measure luminescence signals of singlet oxygen (1 O2 ) produced during photodynamic therapy (PDT) using a CW (continuous wave) light source. The system is based on a compact, fiber-coupled, high collection efficiency spectrometer (>50% transmittance) designed to maximize optical throughput but with sufficient spectral resolution (~7 nm). This is adequate to detect 1 O2 phosphorescence in the presence of strong luminescence background in vivo. This system provides simultaneous acquisition of multiple spectral data points, allowing for more accurate determination of luminescence baseline via spectral fitting and thus the extraction of 1 O2 phosphorescence signal based solely on spectroscopic decomposition, without the need for time-gating. Simultaneous collection of photons at different wavelengths improves the quantum efficiency of the system when compared to sequential spectral measurements such as filter-wheel or tunable-filter based systems. A prototype system was tested during in vivo PDT tumor regression experiments using benzoporphyrin derivative (BPD) photosensitizer. It was found that the treatment efficacy (tumor growth inhibition rate) correlated more strongly with 1 O2 phosphorescence than with PS fluorescence. These results indicate that this high photon-collection efficiency spectrometer instrument may offer a viable option for real-time 1 O2 dosimetry during PDT treatment using CW light.


Subject(s)
Photochemotherapy , Singlet Oxygen , Luminescence , Photosensitizing Agents , Radiation Dosimeters
18.
Int J Radiat Oncol Biol Phys ; 111(1): 240-248, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33845146

ABSTRACT

PURPOSE: Delivery of radiation at ultrahigh dose rates (UHDRs), known as FLASH, has recently been shown to preferentially spare normal tissues from radiation damage compared with tumor tissues. However, the underlying mechanism of this phenomenon remains unknown, with one of the most widely considered hypotheses being that the effect is related to substantial oxygen depletion upon FLASH, thereby altering the radiochemical damage during irradiation, leading to different radiation responses of normal and tumor cells. Testing of this hypothesis would be advanced by direct measurement of tissue oxygen in vivo during and after FLASH irradiation. METHODS AND MATERIALS: Oxygen measurements were performed in vitro and in vivo using the phosphorescence quenching method and a water-soluble molecular probe Oxyphor 2P. The changes in oxygen per unit dose (G-values) were quantified in response to irradiation by 10 MeV electron beam at either UHDR reaching 300 Gy/s or conventional radiation therapy dose rates of 0.1 Gy/s. RESULTS: In vitro experiments with 5% bovine serum albumin solutions at 23°C resulted in G-values for oxygen consumption of 0.19 to 0.21 mm Hg/Gy (0.34-0.37 µM/Gy) for conventional irradiation and 0.16 to 0.17 mm Hg/Gy (0.28-0.30 µM/Gy) for UHDR irradiation. In vivo, the total decrease in oxygen after a single fraction of 20 Gy FLASH irradiation was 2.3 ± 0.3 mm Hg in normal tissue and 1.0 ± 0.2 mm Hg in tumor tissue (P < .00001), whereas no decrease in oxygen was observed from a single fraction of 20 Gy applied in conventional mode. CONCLUSIONS: Our observations suggest that oxygen depletion to radiologically relevant levels of hypoxia is unlikely to occur in bulk tissue under FLASH irradiation. For the same dose, FLASH irradiation induces less oxygen consumption than conventional irradiation in vitro, which may be related to the FLASH sparing effect. However, the difference in oxygen depletion between FLASH and conventional irradiation could not be quantified in vivo because measurements of oxygen depletion under conventional irradiation are hampered by resupply of oxygen from the blood.


Subject(s)
Neoplasms, Experimental/radiotherapy , Oxygen/analysis , Animals , Mice , Neoplasms, Experimental/metabolism , Oxygen Consumption , Radiotherapy Dosage
19.
Photodiagnosis Photodyn Ther ; 30: 101790, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32344195

ABSTRACT

BACKGROUND: Hypoxic lesions often respond poorly to cancer therapies. Particularly, photodynamic therapy (PDT) consumes oxygen in treated tissues, which in turn lowers its efficacy. Tools for online monitoring of intracellular pO2 are desirable. METHODS: The pO2 changes were tracked during photodynamic therapy (PDT) with δ-aminolevulinic acid (ALA) in mouse skin, xenograft tumors, and human skin. ALA was applied either topically as Ameluz cream or systemically by injection. Mitochondrial pO2 was quantified by time-gated lifetime-based imaging of delayed fluorescence (DF) of protoporphyrin IX (PpIX). RESULTS: pO2-weighted images were obtained with capture-times of several seconds, radiant exposures near 10 mJ/cm2, spatial resolution of 0.3 mm, and a broad dynamic range 1-50 mmHg, corresponding to DF lifetimes ≈20-2000 µs. The dose-rate effect on oxygen consumption was investigated in mouse skin. A fluence rate of 1.2 mW/cm2 did not cause any appreciable oxygen depletion, whereas 6 mW/cm2 and 12 mW/cm2 caused severe oxygen depletion after radiant exposures of only 0.4-0.8 J/cm2 and <0.2 J/cm2, respectively. Reoxygenation after PDT was studied too. With a 5 J/cm2 radiant exposure, the recovery times were 10-60 min, whereas with 2 J/cm2 they were only 1-6 min. pO2 distribution was spatially non-uniform at (sub)-millimeter scale, which underlines the necessity of tracking pO2 changes by imaging rather than point-detection. CONCLUSIONS: Time-gated imaging of PpIX DF seems to be a unique tool for direct online monitoring of pO2 changes during PDT with a promising potential for research purposes as well as for comparatively easy clinical translation to improve efficacy in PDT treatment.


Subject(s)
Aminolevulinic Acid/pharmacology , Oxygen Consumption/drug effects , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Protoporphyrins/pharmacokinetics , Aminolevulinic Acid/pharmacokinetics , Animals , Humans , Mice , Mitochondria/drug effects , Optical Imaging , Oxygen/metabolism , Photosensitizing Agents/pharmacokinetics
20.
J Biomed Opt ; 25(6): 1-14, 2020 01.
Article in English | MEDLINE | ID: mdl-31920049

ABSTRACT

Significance: Singlet oxygen is a key cytotoxic agent in photodynamic therapy (PDT). As such, its imaging is highly desirable, but existing direct imaging methods are still limited by the exceptionally low yield of the luminescence signal. Singlet oxygen feedback delayed fluorescence (SOFDF) of the photosensitizer is a higher yield alternative for indirect measurement of this signal.

Aim: The aim was to explore feasibility of SOFDF imaging in vivo in tumor-bearing mice during PDT and investigate how SOFDF images can be transformed into images of singlet oxygen. In addition, we study whether lysosome permeabilization can be visualized through fluorescence lifetime.

Approach: Mice were intravenously injected with 2.5 mg/kg of photosensitizer aluminum(III) phthalocyanine tetrasulfonate (AlPcS4) 20 h prior to experiments, having subcutaneous BxPC3 pancreas tumors. Time-resolved delayed fluorescence and prompt fluorescence (PF) were imaged using an intensified time-gated camera with 10-Hz pulsed laser excitation at 690 nm.

Results: Delayed emission from AlPcS4 was detected with lifetimes 7 to 11 µs, which was attributed to SOFDF and shown to be oxygen-dependent. Singlet oxygen images were approximated by the ratio of SOFDF/PF at each pixel. SOFDF images of a good quality could be captured within several seconds with a radiant exposure of ∼20 mJ / cm2. In addition, lifetime images of AlPcS4 PF in ns-time domain enabled us to visualize the event of lysosome permeabilization, as the lifetime increased from ∼4.7 to 5.2 ns.

Conclusions: Imaging of SOFDF in vivo in mouse tumor during PDT with AlPcS4 is feasible, and it is a promising method for singlet molecular oxygen monitoring. Moreover, the time-gated approach also enables visualization of the lysosome permeabilization that alters the PF lifetime.


Subject(s)
Neoplasms , Photochemotherapy , Animals , Feedback , Fluorescence , Indoles , Lysosomes , Mice , Organometallic Compounds , Photosensitizing Agents/pharmacology , Singlet Oxygen
SELECTION OF CITATIONS
SEARCH DETAIL