Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
2.
Nat Protoc ; 18(3): 990-1015, 2023 03.
Article in English | MEDLINE | ID: mdl-36494493

ABSTRACT

X-ray computed tomography is a reliable technique for the detection and longitudinal monitoring of pulmonary nodules. In preclinical stages of diagnostic or therapeutic development, the miniaturized versions of the clinical computed tomography scanners are ideally suited for carrying out translationally-relevant research in conditions that closely mimic those found in the clinic. In this Protocol, we provide image acquisition parameters optimized for low radiation dose, high-resolution and high-throughput computed tomography imaging using three commercially available micro-computed tomography scanners, together with a detailed description of the image analysis tools required to identify a variety of lung tumor types, characterized by specific radiological features. For each animal, image acquisition takes 4-8 min, and data analysis typically requires 10-30 min. Researchers with basic training in animal handling, medical imaging and software analysis should be able to implement this protocol across a wide range of lung cancer models in mice for investigating the molecular mechanisms driving lung cancer development and the assessment of diagnostic and therapeutic agents.


Subject(s)
Lung Neoplasms , Mice , Animals , X-Ray Microtomography/methods , Lung Neoplasms/diagnostic imaging , Software , Image Processing, Computer-Assisted
3.
Biochem J ; 479(13): 1467-1486, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35730579

ABSTRACT

The protein kinase PKN2 is required for embryonic development and PKN2 knockout mice die as a result of failure in the expansion of mesoderm, cardiac development and neural tube closure. In the adult, cardiomyocyte PKN2 and PKN1 (in combination) are required for cardiac adaptation to pressure-overload. The specific role of PKN2 in contractile cardiomyocytes during development and its role in the adult heart remain to be fully established. We used mice with cardiomyocyte-directed knockout of PKN2 or global PKN2 haploinsufficiency to assess cardiac development and function using high resolution episcopic microscopy, MRI, micro-CT and echocardiography. Biochemical and histological changes were also assessed. Cardiomyocyte-directed PKN2 knockout embryos displayed striking abnormalities in the compact myocardium, with frequent myocardial clefts and diverticula, ventricular septal defects and abnormal heart shape. The sub-Mendelian homozygous knockout survivors developed cardiac failure. RNASeq data showed up-regulation of PKN2 in patients with dilated cardiomyopathy, suggesting an involvement in adult heart disease. Given the rarity of homozygous survivors with cardiomyocyte-specific deletion of PKN2, the requirement for PKN2 in adult mice was explored using the constitutive heterozygous PKN2 knockout. Cardiac hypertrophy resulting from hypertension induced by angiotensin II was reduced in these haploinsufficient PKN2 mice relative to wild-type littermates, with suppression of cardiomyocyte hypertrophy and cardiac fibrosis. It is concluded that cardiomyocyte PKN2 is essential for heart development and the formation of compact myocardium and is also required for cardiac hypertrophy in hypertension. Thus, PKN signalling may offer therapeutic options for managing congenital and adult heart diseases.


Subject(s)
Cardiomyopathies , Hypertension , Protein Kinase C/metabolism , Angiotensin II/metabolism , Angiotensin II/pharmacology , Animals , Cardiomegaly/metabolism , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Female , Hypertension/metabolism , Hypertension/pathology , Mice , Mice, Knockout , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Pregnancy
4.
Methods Mol Biol ; 2366: 321-342, 2021.
Article in English | MEDLINE | ID: mdl-34236648

ABSTRACT

Enforced activation of NF-κB signaling can be achieved by constitutive NF-κB-inducing kinases, IKK2 and NIK, or via lymphoma-associated mutants of MYD88, CARD11, and CD79B. In order to model Diffuse Large B Cell Lymphoma (DLBCL) in mice, conditional alleles for these proteins are combined with alleles targeting Cre recombinase expression in mature B cells. However, unopposed NF-κB signaling promotes plasmablast differentiation, and as a consequence the model system must be complemented with further mutations that block differentiation, such as Prdm1/BLIMP1 inactivation or overexpression of BCL6. Here, we describe the currently available tools for DLBCL models in mice and their relative advantages and drawbacks. Furthermore, we describe methods to monitor lymphomagenesis, using ultrasound tomography of the spleen, and the technique of partial splenectomy surgery with recovery. These powerful techniques allow paired comparison of individual lymphoma cases before and after interventions, including therapies, and to study the evolution of lymphoma over time. NF-κB activation also promotes widespread nodal involvement with lymphoma and we describe the post-mortem dissection of major nodal groups.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Animals , B-Lymphocytes/metabolism , Disease Models, Animal , Lymphoma, Large B-Cell, Diffuse/genetics , Mice , Mutation , NF-kappa B/genetics , NF-kappa B/metabolism , Signal Transduction
5.
Blood Cancer Discov ; 2(2): 135-145, 2021 03.
Article in English | MEDLINE | ID: mdl-33778768

ABSTRACT

Myelodysplastic syndrome (MDS) are clonal stem cell diseases characterized mainly by ineffective hematopoiesis. Here, we present an approach that enables robust long-term engraftment of primary MDS stem cells (MDS-SCs) in mice by implantation of human mesenchymal cell-seeded scaffolds. Critically for modelling MDS, where patient sample material is limiting, mononuclear bone marrow cells containing as few as 104 CD34+ cells can be engrafted and expanded by this approach with the maintenance of the genetic make-up seen in the patients. Non-invasive high-resolution ultrasound imaging shows that these scaffolds are fully perfused. Our data shows that human microenvironment but not mouse is essential to MDS-SCs homing and engraftment. Notably, the alternative niche provided by healthy donor MSCs enhanced engraftment of MDS-SCs. This study characterizes a new tool to model MDS human disease with the level of engraftment previously unattainable in mice, and offers insights into human-specific determinants of MDS-SC microenvironment.


Subject(s)
Mesenchymal Stem Cells , Myelodysplastic Syndromes , Animals , Bone Marrow Cells , Hematopoiesis , Humans , Mice , Stem Cells
6.
Development ; 147(21)2020 11 12.
Article in English | MEDLINE | ID: mdl-32994166

ABSTRACT

The Hippo-YAP/TAZ pathway is an important regulator of tissue growth, but can also control cell fate or tissue morphogenesis. Here, we investigate the function of the Hippo pathway during the development of cartilage, which forms the majority of the skeleton. Previously, YAP was proposed to inhibit skeletal size by repressing chondrocyte proliferation and differentiation. We find that, in vitro, Yap/Taz double knockout impairs murine chondrocyte proliferation, whereas constitutively nuclear nls-YAP5SA accelerates proliferation, in line with the canonical role of this pathway in most tissues. However, in vivo, cartilage-specific knockout of Yap/Taz does not prevent chondrocyte proliferation, differentiation or skeletal growth, but rather results in various skeletal deformities including cleft palate. Cartilage-specific expression of nls-YAP5SA or knockout of Lats1/2 do not increase cartilage growth, but instead lead to catastrophic malformations resembling chondrodysplasia or achondrogenesis. Physiological YAP target genes in cartilage include Ctgf, Cyr61 and several matrix remodelling enzymes. Thus, YAP/TAZ activity controls chondrocyte proliferation in vitro, possibly reflecting a regenerative response, but is dispensable for chondrocyte proliferation in vivo, and instead functions to control cartilage morphogenesis via regulation of the extracellular matrix.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bone and Bones/embryology , Bone and Bones/metabolism , Cell Cycle Proteins/metabolism , Morphogenesis , Protein Serine-Threonine Kinases/metabolism , Trans-Activators/metabolism , Animals , Bone and Bones/abnormalities , Bone and Bones/pathology , Cartilage/pathology , Cell Nucleus/metabolism , Cell Proliferation , Chondrocytes/metabolism , Chondrocytes/pathology , Cleft Palate/pathology , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Gene Expression Regulation, Developmental , Growth Plate/pathology , Hippo Signaling Pathway , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis/genetics , Signal Transduction , Tumor Suppressor Proteins/metabolism , YAP-Signaling Proteins
7.
J Clin Med ; 8(12)2019 12 11.
Article in English | MEDLINE | ID: mdl-31835761

ABSTRACT

A cogent issue in cancer research is how to account for the effects of tumor microenvironment (TME) on the response to therapy, warranting the need to adopt adequate in vitro and in vivo models. This is particularly relevant in the development of strategies targeting cancer metabolism, as they will inevitably have systemic effects. For example, inhibition of mitochondrial complex I (CI), despite showing promising results as an anticancer approach, triggers TME-mediated survival mechanisms in subcutaneous osteosarcoma xenografts, a response that may vary according to whether the tumors are induced via subcutaneous injection or by intrabone orthotopic transplantation. Thus, with the aim to characterize the TME of CI-deficient tumors in a model that more faithfully represents osteosarcoma development, we set up a humanized bone niche ectopic graft. A prominent involvement of TME was revealed in CI-deficient tumors, characterized by the abundance of cancer associated fibroblasts, tumor associated macrophages and preservation of osteocytes and osteoblasts in the mineralized bone matrix. The pseudo-orthotopic approach allowed investigation of osteosarcoma progression in a bone-like microenvironment setting, without being invasive as the intrabone cell transplantation. Additionally, establishing osteosarcomas in a humanized bone niche model identified a peculiar association between targeting CI and bone tissue preservation.

8.
Front Oncol ; 6: 221, 2016.
Article in English | MEDLINE | ID: mdl-27818949

ABSTRACT

PURPOSE: Most effective antitumor therapies induce tumor cell death. Non-invasive, rapid and accurate quantitative imaging of cell death is essential for monitoring early response to antitumor therapies. To facilitate this, we previously developed a biocompatible necrosis-avid near-infrared fluorescence (NIRF) imaging probe, HQ4, which was radiolabeled with 111Indium-chloride (111In-Cl3) via the chelate diethylene triamine pentaacetic acid (DTPA), to enable clinical translation. The aim of the present study was to evaluate the application of HQ4-DTPA for monitoring tumor cell death induced by radiation therapy. Apart from its NIRF and radioactive properties, HQ4-DTPA was also tested as a photoacoustic imaging probe to evaluate its performance as a multimodal contrast agent for superficial and deep tissue imaging. MATERIALS AND METHODS: Radiation-induced tumor cell death was examined in a xenograft mouse model of human breast cancer (MCF-7). Tumors were irradiated with three fractions of 9 Gy each. HQ4-DTPA was injected intravenously after the last irradiation, NIRF and photoacoustic imaging of the tumors were performed at 12, 20, and 40 h after injection. Changes in probe accumulation in the tumors were measured in vivo, and ex vivo histological analysis of excised tumors was performed at experimental endpoints. In addition, biodistribution of radiolabeled [111In]DTPA-HQ4 was assessed using hybrid single-photon emission computed tomography-computed tomography (SPECT-CT) at the same time points. RESULTS: In vivo NIRF imaging demonstrated a significant difference in probe accumulation between control and irradiated tumors at all time points after injection. A similar trend was observed using in vivo photoacoustic imaging, which was validated by ex vivo tissue fluorescence and photoacoustic imaging. Serial quantitative radioactivity measurements of probe biodistribution further demonstrated increased probe accumulation in irradiated tumors. CONCLUSION: HQ4-DTPA has high specificity for dead cells in vivo, potentiating its use as a contrast agent for determining the relative level of tumor cell death following radiation therapy using NIRF, photoacoustic imaging and SPECT in vivo. Initial preclinical results are promising and indicate the need for further evaluation in larger cohorts. If successful, such studies may help develop a new multimodal method for non-invasive and dynamic deep tissue imaging of treatment-induced cell death to quantitatively assess therapeutic response in patients.

9.
Mol Imaging Biol ; 18(6): 905-915, 2016 12.
Article in English | MEDLINE | ID: mdl-27277828

ABSTRACT

PURPOSE: Recently we showed that a number of carboxylated near-infrared fluorescent (NIRF) cyanine dyes possess strong necrosis avid properties in vitro as well as in different mouse models of spontaneous and therapy-induced tumor necrosis, indicating their potential use for cancer diagnostic- and prognostic purposes. In the previous study, the detection of the cyanines was achieved by whole body optical imaging, a technique that, due to the limited penetration of near-infrared light, is not suitable for investigations deeper than 1 cm within the human body. Therefore, in order to facilitate clinical translation, the purpose of the present study was to generate a necrosis avid cyanine-based NIRF probe that could also be used for single photon emission computed tomography (SPECT). For this, the necrosis avid NIRF cyanine HQ4 was radiolabeled with 111indium, via the chelate diethylene triamine pentaacetic acid (DTPA). PROCEDURES: The necrosis avid properties of the radiotracer [111In]DTPA-HQ4 were examined in vitro and in vivo in different breast tumor models in mice using SPECT and optical imaging. Moreover, biodistribution studies were performed to examine the pharmacokinetics of the probe in vivo. RESULTS: Using optical imaging and radioactivity measurements, in vitro, we showed selective accumulation of [111In]DTPA-HQ4 in dead cells. Using SPECT and in biodistribution studies, the necrosis avidity of the radiotracer was confirmed in a 4T1 mouse breast cancer model of spontaneous tumor necrosis and in a MCF-7 human breast cancer model of chemotherapy-induced tumor necrosis. CONCLUSIONS: The radiotracer [111In]DTPA-HQ4 possessed strong and selective necrosis avidity in vitro and in various mouse models of tumor necrosis in vivo, indicating its potential to be clinically applied for diagnostic purposes and to monitor anti-cancer treatment efficacy.


Subject(s)
Carbocyanines/chemistry , Multimodal Imaging/methods , Neoplasms/diagnostic imaging , Neoplasms/pathology , Tomography, Emission-Computed, Single-Photon/methods , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Indium Radioisotopes/chemistry , Mice, Inbred BALB C , Mice, Nude , Necrosis , Optical Imaging , Pentetic Acid/chemistry , Tissue Distribution
10.
J Control Release ; 229: 93-105, 2016 05 10.
Article in English | MEDLINE | ID: mdl-26988602

ABSTRACT

Photodynamic therapy (PDT) induces cell death through local light activation of a photosensitizer (PS) and has been used to treat head and neck cancers. Yet, common PS lack tumor specificity, which leads to collateral damage to normal tissues. Targeted delivery of PS via antibodies has pre-clinically improved tumor selectivity. However, antibodies have long half-lives and relatively poor tissue penetration, which could limit therapeutic efficacy and lead to long photosensitivity. Here, in this feasibility study, we evaluate at the pre-clinical level a recently introduced format of targeted PDT, which employs nanobodies as targeting agents and a water-soluble PS (IRDye700DX) that is traceable through optical imaging. In vitro, the PS solely binds to cells and induces phototoxicity on cells overexpressing the epidermal growth factor receptor (EGFR), when conjugated to the EGFR targeted nanobodies. To investigate whether this new format of targeted PDT is capable of inducing selective tumor cell death in vivo, PDT was applied on an orthotopic mouse tumor model with illumination at 1h post-injection of the nanobody-PS conjugates, as selected from quantitative fluorescence spectroscopy measurements. In parallel, and as a reference, PDT was applied with an antibody-PS conjugate, with illumination performed 24h post-injection. Importantly, EGFR targeted nanobody-PS conjugates led to extensive tumor necrosis (approx. 90%) and almost no toxicity in healthy tissues, as observed through histology 24h after PDT. Overall, results show that these EGFR targeted nanobody-PS conjugates are selective and able to induce tumor cell death in vivo. Additional studies are now needed to assess the full potential of this approach to improving PDT.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , ErbB Receptors/metabolism , Indoles/administration & dosage , Organosilicon Compounds/administration & dosage , Photochemotherapy , Photosensitizing Agents/administration & dosage , Single-Domain Antibodies/administration & dosage , Tongue Neoplasms/drug therapy , Animals , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Coculture Techniques , Female , Humans , Indoles/therapeutic use , Light , Mice, Inbred BALB C , Mice, Nude , Organosilicon Compounds/therapeutic use , Photosensitizing Agents/therapeutic use , Single-Domain Antibodies/therapeutic use , Tongue Neoplasms/metabolism
11.
Clin Cancer Res ; 22(6): 1459-68, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26546617

ABSTRACT

PURPOSE: The efficacy of immunotherapy against advanced cancer may be improved by combination strategies. Photodynamic therapy (PDT) is a local tumor ablation method based on localized activation of a photosensitizer, leading to oxygen radical-induced tumor cell death. PDT can enhance antitumor immune responses by release of antigen and danger signals, supporting combination protocols of PDT with immunotherapy. EXPERIMENTAL DESIGN: We investigated the local and systemic immune effects of PDT after treatment of established tumors. In two independent aggressive mouse tumor models, TC-1 and RMA, we combined PDT with therapeutic vaccination using synthetic long peptides (SLP) containing epitopes from tumor antigens. RESULTS: PDT of established tumors using the photosensitizer Bremachlorin resulted in significant delay of tumor outgrowth. Combination treatment of PDT with therapeutic SLP vaccination cured one third of mice. Importantly, all cured mice were fully protected against subsequent tumor rechallenge, and combination treatment of primary tumors led to eradication of distant secondary tumors, indicating the induction of a systemic antitumor immune response. Indeed, PDT by itself induced a significant CD8(+) T-cell response against the tumor, which was increased when combined with SLP vaccination and essential for the therapeutic effect of combination therapy. CONCLUSIONS: We show that immunotherapy can be efficiently combined with PDT to eradicate established tumors, based on strong local tumor ablation and the induction of a robust systemic immune response. These results suggest combination of active immunotherapy with tumor ablation by PDT as a feasible novel treatment strategy for advanced cancer.


Subject(s)
Immunotherapy , Neoplasms/immunology , Neoplasms/pathology , Photochemotherapy , Animals , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/immunology , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Female , Humans , Immunomodulation , Immunotherapy/methods , Mice , Neoplasms/mortality , Neoplasms/therapy , Photosensitizing Agents/pharmacology , Tumor Burden/drug effects , Tumor Burden/immunology , Vaccines, Subunit/immunology
12.
J Control Release ; 223: 31-41, 2016 Feb 10.
Article in English | MEDLINE | ID: mdl-26708021

ABSTRACT

Necrotic cell death occurs exclusively under pathological conditions, such as ischemic diseases. Necrosis imaging is of diagnostic value and enables early measurement of treatment efficiency in ischemic patients. Here we explored the targeted delivery of particles, with diameters of approximately 100nm, 200nm and 800nm, consisting of a poly(lactic-co-glycolic acid) (PLGA) nanoparticle (NP) core coated with a polyethylene glycol-lipid (PEG) layer. Targeted delivery was facilitated by coupling the amino end group of the polyethylene glycol-layer to 800CW imaging agent, which specifically binds to intracellular proteins of cells that have lost membrane integrity, thus revealing the extent of the damaged area. We found that smaller NPs (100nm), with an appropriate coating, diffuse throughout the traumatic brain injury (TBI) in mice. Optical imaging revealed that smaller (100-nm) PEG-coated NPs carrying 800CW penetrated deeper into the mouse brain than large 800CW containing NPs (800nm). The importance of the 800CW as a ligand to target the necrotic tissue was further confirmed in living mice. The ability to achieve brain penetration with smaller NPs is expected to allow more uniform, longer-lasting, and effective delivery of drugs within the brain, and may find application in the treatment of stroke, brain tumors, neuroinflammation, and other brain diseases where the blood-brain barrier is compromised or where local delivery strategies are feasible.


Subject(s)
Brain Injuries/metabolism , Brain/metabolism , Drug Carriers , Lactic Acid , Nanoparticles , Polyglycolic Acid , Animals , Benzenesulfonates/administration & dosage , Benzenesulfonates/pharmacokinetics , Cell Line, Tumor , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Female , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/pharmacokinetics , Indoles/administration & dosage , Indoles/pharmacokinetics , Lactic Acid/administration & dosage , Lactic Acid/chemistry , Lactic Acid/pharmacokinetics , Mice , Mice, Inbred BALB C , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Particle Size , Polyglycolic Acid/administration & dosage , Polyglycolic Acid/chemistry , Polyglycolic Acid/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer , Tissue Distribution
13.
Oncotarget ; 6(36): 39036-49, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26472022

ABSTRACT

Quantification of tumor necrosis in cancer patients is of diagnostic value as the amount of necrosis is correlated with disease prognosis and it could also be used to predict early efficacy of anti-cancer treatments. In the present study, we identified two near infrared fluorescent (NIRF) carboxylated cyanines, HQ5 and IRDye 800CW (800CW), which possess strong necrosis avidity. In vitro studies showed that both dyes selectively bind to cytoplasmic proteins of dead cells that have lost membrane integrity. Affinity for cytoplasmic proteins was confirmed using quantitative structure activity relations modeling. In vivo results, using NIRF and optoacoustic imaging, confirmed the necrosis avid properties of HQ5 and 800CW in a mouse 4T1 breast cancer tumor model of spontaneous necrosis. Finally, in a mouse EL4 lymphoma tumor model, already 24 h post chemotherapy, a significant increase in 800CW fluorescence intensity was observed in treated compared to untreated tumors. In conclusion, we show, for the first time, that the NIRF carboxylated cyanines HQ5 and 800CW possess strong necrosis avid properties in vitro and in vivo. When translated to the clinic, these dyes may be used for diagnostic or prognostic purposes and for monitoring in vivo tumor response early after the start of treatment.


Subject(s)
Carbocyanines/chemistry , Fluorescent Dyes/chemistry , Lymphoma/diagnostic imaging , Lymphoma/drug therapy , Mammary Neoplasms, Experimental/diagnostic imaging , Mammary Neoplasms, Experimental/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Death/physiology , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Lymphoma/pathology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Confocal/methods , Necrosis/pathology , Quantitative Structure-Activity Relationship , Random Allocation
14.
J Pathol ; 235(5): 745-59, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25421310

ABSTRACT

Invasive ductal carcinoma (IDC) and invasive lobular carcinoma (ILC) are the most frequently occurring histological subtypes of breast cancer, accounting for 80-90% and 10-15% of the total cases, respectively. At the time of diagnosis and surgical resection of the primary tumour, most patients do not have clinical signs of metastases, but bone micrometastases may already be present. Our aim was to develop a novel preclinical ILC model of spontaneous bone micrometastasis. We used murine invasive lobular breast carcinoma cells (KEP) that were generated by targeted deletion of E-cadherin and p53 in a conditional K14cre;Cdh1((F/F));Trp53((F/F)) mouse model of de novo mammary tumour formation. After surgical resection of the growing orthotopically implanted KEP cells, distant metastases were formed. In contrast to other orthotopic breast cancer models, KEP cells readily formed skeletal metastases with minimal lung involvement. Continuous treatment with SD-208 (60 mg/kg per day), an orally available TGFß receptor I kinase inhibitor, increased the tumour growth at the primary site and increased the number of distant metastases. Furthermore, when SD-208 treatment was started after surgical resection of the orthotopic tumour, increased bone colonisation was also observed (versus vehicle). Both our in vitro and in vivo data show that SD-208 treatment reduced TGFß signalling, inhibited apoptosis, and increased proliferation. In conclusion, we have demonstrated that orthotopic implantation of murine ILC cells represent a new breast cancer model of minimal residual disease in vivo, which comprises key steps of the metastatic cascade. The cancer cells are sensitive to the anti-tumour effects of TGFß. Our in vivo model is ideally suited for functional studies and evaluation of new pharmacological intervention strategies that may target one or more steps along the metastatic cascade of events.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/pathology , Carcinoma, Lobular/secondary , Mammary Neoplasms, Experimental/pathology , Protein Kinase Inhibitors/toxicity , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pteridines/toxicity , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Animals , Apoptosis/drug effects , Bone Neoplasms/enzymology , Bone Neoplasms/genetics , Breast Neoplasms/chemically induced , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Carcinoma, Lobular/chemically induced , Carcinoma, Lobular/enzymology , Carcinoma, Lobular/genetics , Cdh1 Proteins/deficiency , Cdh1 Proteins/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/genetics , Mice, Knockout , Neoplasm Micrometastasis , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Time Factors , Transfection , Tumor Burden/drug effects , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
15.
PLoS One ; 9(9): e108730, 2014.
Article in English | MEDLINE | ID: mdl-25265510

ABSTRACT

In small animal imaging studies, when the locations of the micro-structures of interest are unknown a priori, there is a simultaneous need for full-body coverage and high resolution. In MRI, additional requirements to image contrast and acquisition time will often make it impossible to acquire such images directly. Recently, a resolution enhancing post-processing technique called super-resolution reconstruction (SRR) has been demonstrated to improve visualization and localization of micro-structures in small animal MRI by combining multiple low-resolution acquisitions. However, when the field-of-view is large relative to the desired voxel size, solving the SRR problem becomes very expensive, in terms of both memory requirements and computation time. In this paper we introduce a novel local approach to SRR that aims to overcome the computational problems and allow researchers to efficiently explore both global and local characteristics in whole-body small animal MRI. The method integrates state-of-the-art image processing techniques from the areas of articulated atlas-based segmentation, planar reformation, and SRR. A proof-of-concept is provided with two case studies involving CT, BLI, and MRI data of bone and kidney tumors in a mouse model. We show that local SRR-MRI is a computationally efficient complementary imaging modality for the precise characterization of tumor metastases, and that the method provides a feasible high-resolution alternative to conventional MRI.


Subject(s)
Bone Neoplasms/secondary , Kidney Neoplasms/secondary , Magnetic Resonance Imaging , Animals , Bone Neoplasms/diagnostic imaging , Image Processing, Computer-Assisted , Kidney Neoplasms/diagnostic imaging , Luminescent Measurements , Mice, Inbred BALB C , Phantoms, Imaging , Pilot Projects , Time Factors , Tomography, X-Ray Computed , Whole Body Imaging
16.
J Biomed Opt ; 19(1): 15010, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24477382

ABSTRACT

Quantification of fluorescence in vivo is complicated by the influence of tissue optical properties on the collected fluorescence signal. When tissue optical properties in the measurement volume are quantified, one can obtain the intrinsic fluorescence, which equals the product of fluorophore absorption coefficient and quantum yield. We applied this method to in vivo single-fiber fluorescence spectroscopy measurements on mouse tongue, skin, liver, and oral squamous cell carcinoma, where we detected intrinsic fluorescence spectra of the photosensitizers chlorin e6 and Bremachlorin at t=[3,4.5,6,24,48] h incubation time. We observed a tissue-dependent maximum of 35% variation in the total correction factor over the visible wavelength range. Significant differences in spectral shape over time between sensitizers were observed. Although the wavelength position of the fluorescence intensity maximum for ce6 shifted to the red, Bremachlorin showed a blue shift. Furthermore, the Bremachlorin peak appeared to be broader than the ce6 fluorescence peak. Intrinsic fluorescence intensity, which can be related to photosensitizer concentration, was decreasing for all time points but showed significantly more Bremachlorin present compared to ce6 at long incubation times. Results from this study can be used to define an optimal treatment protocol for Bremachlorin-based photodynamic therapy.


Subject(s)
Chlorophyll/analogs & derivatives , Photosensitizing Agents/chemistry , Animals , Carcinoma, Squamous Cell/pathology , Chlorophyll/chemistry , Chlorophyllides , Female , Fluorescence , Green Fluorescent Proteins , Liver/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Fluorescence , Mouth Neoplasms/pathology , Normal Distribution , Optics and Photonics , Photochemotherapy , Porphyrins/chemistry , Skin/pathology , Spectrometry, Fluorescence , Spectrophotometry , Tongue/pathology
17.
Lasers Surg Med ; 46(3): 224-34, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24436109

ABSTRACT

BACKGROUND AND OBJECTIVE: The effect of photodynamic therapy (PDT) is dependent on the localization of photosensitizer in the treatment volume at the time of illumination. Investigation of photosensitizer pharmacokinetics in and around the treatment volume aids in determining the optimal drug light interval for PDT. MATERIALS AND METHODS: In this paper we have investigated the distribution of the photosensitizers chlorin e6 and Bremachlorin in the oral squamous cell carcinoma cell-line OSC19-Luc-Gfp in a tongue tumor, tumor boundary, invasive tumor boundary, and normal tongue tissue by the use of confocal microscopy of frozen sections. Tongues were harvested at t = [3, 4.5, 6, 24, 48] hours after injection. RESULTS: Both photosensitizers showed a decreasing fluorescence with increasing incubation time, and at all time points higher fluorescence was measured in tumor boundary than in tumor itself. For short incubation times, a higher fluorescence intensity was observed in the invasive tumor border and normal tissue compared to tumor tissue. Bremachlorin showed a small increase in tumor to normal ratio at 24 and 48 hours incubation time. Ce6 was undetectable at 48 hours. We did not find a correlation between photosensitizer localization and the presence of vasculature. CONCLUSION: The modest tumor/tumor boundary to normal selectivity of between 1.2 and 2.5 exhibited by Bremachlorin 24 and 48 hours after administration may allow selective targeting of tongue tumors. Further studies investigating the relationship between Bremachlorin concentration and therapeutic efficacy PDT with long incubation times are warranted.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Photochemotherapy , Photosensitizing Agents/pharmacokinetics , Porphyrins/pharmacokinetics , Tongue Neoplasms/drug therapy , Animals , Chlorophyllides , Drug Combinations , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Photosensitizing Agents/therapeutic use , Porphyrins/therapeutic use , Random Allocation
18.
Clin Cancer Res ; 19(14): 3745-54, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23674494

ABSTRACT

Optical image-guided cancer surgery is a promising technique to adequately determine tumor margins by tumor-specific targeting, potentially resulting in complete resection of tumor tissue with improved survival. However, identification of the photons coming from the fluorescent contrast agent is complicated by autofluorescence, optical tissue properties, and accurate fluorescent targeting agents and imaging systems. All these factors have an important influence on the image that is presented to the surgeon. Considering the clinical consequences at stake, it is a prerequisite to answer the questions that are essential for the surgeon. What is optical image-guided surgery and how can it improve patient care? What should the oncologic surgeon know about the fundamental principles of optical imaging to understand which conclusions can be drawn from the images? And how do the limitations influence clinical decision making? This article discusses these questions and provides a clear overview of the basic principles and practical applications. Although there are limitations to the intrinsic capacity of the technique, when practical and technical surgical possibilities are considered, optical imaging can be a very powerful intraoperative tool in guiding the future oncologic surgeon toward radical resection and optimal clinical results.


Subject(s)
Neoplasms/surgery , Surgery, Computer-Assisted , Animals , Humans , Neoplasms/pathology , Optical Imaging/methods , Scattering, Radiation
19.
PLoS One ; 7(2): e31875, 2012.
Article in English | MEDLINE | ID: mdl-22348134

ABSTRACT

Bioluminescence imaging (BLI) has shown its appeal as a sensitive technique for in vivo whole body optical imaging. However, the development of injectable tumor-specific near-infrared fluorescent (NIRF) probes makes fluorescence imaging (FLI) a promising alternative to BLI in situations where BLI cannot be used or is unwanted (e.g., spontaneous transgenic tumor models, or syngeneic mice to study immune effects).In this study, we addressed the questions whether it is possible to detect tumor progression using FLI with appropriate sensitivity and how FLI correlates with BLI measurements. In addition, we explored the possibility to simultaneously detect multiple tumor characteristics by dual-wavelength FLI (~700 and ~800 nm) in combination with spectral unmixing. Using a luciferase-expressing 4T1-luc2 mouse breast cancer model and combinations of activatable and targeting NIRF probes, we showed that the activatable NIRF probes (ProSense680 and MMPSense680) and the targeting NIRF probes (IRDye 800CW 2-DG and IRDye 800CW EGF) were either activated by or bound to 4T1-luc2 cells. In vivo, we implanted 4T1-luc2 cells orthotopically in nude mice and were able to follow tumor progression longitudinally both by BLI and dual-wavelength FLI. We were able to reveal different probe signals within the tumor, which co-localized with immuno-staining. Moreover, we observed a linear correlation between the internal BLI signals and the FLI signals obtained from the NIRF probes. Finally, we could detect pulmonary metastases both by BLI and FLI and confirmed their presence histologically.Taken together, these data suggest that dual-wavelength FLI is a feasible approach to simultaneously detect different features of one tumor and to follow tumor progression with appropriate specificity and sensitivity. This study may open up new perspectives for the detection of tumors and metastases in various experimental models and could also have clinical applications, such as image-guided surgery.


Subject(s)
Diagnostic Imaging/methods , Fluorescent Dyes , Luminescent Measurements/methods , Mammary Neoplasms, Experimental/diagnosis , Animals , Benzenesulfonates , Diagnostic Imaging/instrumentation , Disease Models, Animal , Disease Progression , Indoles , Luminescent Measurements/instrumentation , Mammary Neoplasms, Experimental/pathology , Mice
20.
Methods Mol Biol ; 816: 507-15, 2012.
Article in English | MEDLINE | ID: mdl-22130947

ABSTRACT

Optical imaging is a valuable technique for visualizing and quantifying biological processes in living -organisms. Optical imaging can be divided into two main imaging modalities: bioluminescence imaging and fluorescence imaging. This chapter describes the use of these imaging techniques to image tumour cells in mouse models of cancer and to detect early bone metastasis.


Subject(s)
Bone and Bones/pathology , Diagnostic Imaging/methods , Luminescent Measurements/methods , Neoplasm Metastasis/diagnosis , Neoplasms/diagnosis , Animals , Mice , Neoplasm Metastasis/pathology , Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...