Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
2.
Magn Reson Med ; 90(3): 894-909, 2023 09.
Article in English | MEDLINE | ID: mdl-37093981

ABSTRACT

PURPOSE: To develop a high spatiotemporal resolution 3D dynamic pulse sequence for preclinical imaging of hyperpolarized [1-13 C]pyruvate-to-[1-13 C]lactate metabolism at 7T. METHODS: A standard 3D balanced SSFP (bSSFP) sequence was modified to enable alternating-frequency excitations. RF pulses with 2.33 ms duration and 900 Hz FWHM were placed off-resonance of the target metabolites, [1-13 C]pyruvate (by approximately -245 Hz) and [1-13 C]lactate (by approximately 735 Hz), to selectively excite those resonances. Relatively broad bandwidth (compared to those metabolites' chemical shift offset) permits a short TR of 6.29 ms, enabling higher spatiotemporal resolution. Bloch equation simulations of the bSSFP response profile guided the sequence parameter selection to minimize spectral contamination between metabolites and preserve magnetization over time. RESULTS: Bloch equation simulations, phantom studies, and in vivo studies demonstrated that the two target resonances could be cleanly imaged without substantial bSSFP banding artifacts and with little spectral contamination between lactate and pyruvate and from pyruvate hydrate. High spatiotemporal resolution 3D images were acquired of in vivo pyruvate-lactate metabolism in healthy wild-type and endogenous pancreatic tumor-bearing mice, with 1.212 s acquisition time per single-metabolite image and (1.75 mm)3 isotropic voxels with full mouse abdomen 56 × 28 × 21 mm3 FOV and fully-sampled k-space. Kidney and tumor lactate/pyruvate ratios of two consecutive measurements in one animal, 1 h apart, were consistent. CONCLUSION: Spectrally selective bSSFP using off-resonant RF excitations can provide high spatio-temporal resolution 3D dynamic images of pyruvate-lactate metabolic conversion.


Subject(s)
Lactic Acid , Pyruvic Acid , Mice , Animals , Pyruvic Acid/metabolism , Lactic Acid/metabolism , Magnetic Resonance Imaging/methods , Imaging, Three-Dimensional/methods , Phantoms, Imaging , Carbon Isotopes/metabolism
3.
Cancer Metab ; 10(1): 24, 2022 Dec 09.
Article in English | MEDLINE | ID: mdl-36494842

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) lacks effective treatment options beyond chemotherapy. Although molecular subtypes such as classical and QM (quasi-mesenchymal)/basal-like with transcriptome-based distinct signatures have been identified, deduced therapeutic strategies and targets remain elusive. Gene expression data show enrichment of glycolytic genes in the more aggressive and therapy-resistant QM subtype. However, whether the glycolytic transcripts are translated into functional glycolysis that could further be explored for metabolic targeting in QM subtype is still not known. METHODS: We used different patient-derived PDAC model systems (conventional and primary patient-derived cells, patient-derived xenografts (PDX), and patient samples) and performed transcriptional and functional metabolic analysis. These included RNAseq and Illumina HT12 bead array, in vitro Seahorse metabolic flux assays and metabolic drug targeting, and in vivo hyperpolarized [1-13C]pyruvate and [1-13C]lactate magnetic resonance spectroscopy (HP-MRS) in PDAC xenografts. RESULTS: We found that glycolytic metabolic dependencies are not unambiguously functionally exposed in all QM PDACs. Metabolic analysis demonstrated functional metabolic heterogeneity in patient-derived primary cells and less so in conventional cell lines independent of molecular subtype. Importantly, we observed that the glycolytic product lactate is actively imported into the PDAC cells and used in mitochondrial oxidation in both classical and QM PDAC cells, although more actively in the QM cell lines. By using HP-MRS, we were able to noninvasively identify highly glycolytic PDAC xenografts by detecting the last glycolytic enzymatic step and prominent intra-tumoral [1-13C]pyruvate and [1-13C]lactate interconversion in vivo. CONCLUSION: Our study adds functional metabolic phenotyping to transcriptome-based analysis and proposes a functional approach to identify highly glycolytic PDACs as candidates for antimetabolic therapeutic avenues.

4.
Eur J Nucl Med Mol Imaging ; 50(1): 115-129, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36074156

ABSTRACT

PURPOSE: Pancreatic ductal adenocarcinoma (PDAC) is a molecularly heterogeneous tumor entity with no clinically established imaging biomarkers. We hypothesize that tumor morphology and physiology, including vascularity and perfusion, show variations that can be detected by differences in contrast agent (CA) accumulation measured non-invasively. This work seeks to establish imaging biomarkers for tumor stratification and therapy response monitoring in PDAC, based on this hypothesis. METHODS AND MATERIALS: Regional CA accumulation in PDAC was correlated with tumor vascularization, stroma content, and tumor cellularity in murine and human subjects. Changes in CA distribution in response to gemcitabine (GEM) were monitored longitudinally with computed tomography (CT) Hounsfield Units ratio (HUr) of tumor to the aorta or with magnetic resonance imaging (MRI) ΔR1 area under the curve at 60 s tumor-to-muscle ratio (AUC60r). Tissue analyses were performed on co-registered samples, including endothelial cell proliferation and cisplatin tissue deposition as a surrogate of chemotherapy delivery. RESULTS: Tumor cell poor, stroma-rich regions exhibited high CA accumulation both in human (meanHUr 0.64 vs. 0.34, p < 0.001) and mouse PDAC (meanAUC60r 2.0 vs. 1.1, p < 0.001). Compared to the baseline, in vivo CA accumulation decreased specifically in response to GEM treatment in a subset of human (HUr -18%) and mouse (AUC60r -36%) tumors. Ex vivo analyses of mPDAC showed reduced cisplatin delivery (GEM: 0.92 ± 0.5 mg/g, vs. vehicle: 3.1 ± 1.5 mg/g, p = 0.004) and diminished endothelial cell proliferation (GEM: 22.3% vs. vehicle: 30.9%, p = 0.002) upon GEM administration. CONCLUSION: In PDAC, CA accumulation, which is related to tumor vascularization and perfusion, inversely correlates with tumor cellularity. The standard of care GEM treatment results in decreased CA accumulation, which impedes drug delivery. Further investigation is warranted into potentially detrimental effects of GEM in combinatorial therapy regimens.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Mice , Animals , Cisplatin/therapeutic use , Xenograft Model Antitumor Assays , Carcinoma, Pancreatic Ductal/diagnostic imaging , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Neovascularization, Pathologic/diagnostic imaging , Neovascularization, Pathologic/drug therapy , Biomarkers , Tomography, X-Ray Computed , Magnetic Resonance Imaging , Tomography , Cell Line, Tumor , Gemcitabine , Pancreatic Neoplasms
5.
EJNMMI Res ; 11(1): 83, 2021 Aug 28.
Article in English | MEDLINE | ID: mdl-34453623

ABSTRACT

Correlation of in vivo imaging to histomorphological pathology in animal models requires comparative interdisciplinary expertise of different fields of medicine. From the morphological point of view, there is an urgent need to improve histopathological evaluation in animal model-based research to expedite translation into clinical applications. While different other fields of translational science were standardized over the last years, little was done to improve the pipeline of experimental pathology to ensure reproducibility based on pathological expertise in experimental animal models with respect to defined guidelines and classifications. Additionally, longitudinal analyses of preclinical models often use a variety of imaging methods and much more attention should be drawn to enable for proper co-registration of in vivo imaging methods with the ex vivo morphological read-outs. Here we present the development of the Comparative Experimental Pathology (CEP) unit embedded in the Institute of Pathology of the Technical University of Munich during the Collaborative Research Center 824 (CRC824) funding period together with selected approaches of histomorphological techniques for correlation of in vivo imaging to morphomolecular pathology.

7.
Gastroenterology ; 161(1): 318-332.e9, 2021 07.
Article in English | MEDLINE | ID: mdl-33819482

ABSTRACT

BACKGROUND & AIMS: The existence of different subtypes of pancreatic ductal adenocarcinoma (PDAC) and their correlation with patient outcome have shifted the emphasis on patient classification for better decision-making algorithms and personalized therapy. The contribution of mechanisms regulating the cancer stem cell (CSC) population in different subtypes remains unknown. METHODS: Using RNA-seq, we identified B-cell CLL/lymphoma 3 (BCL3), an atypical nf-κb signaling member, as differing in pancreatic CSCs. To determine the biological consequences of BCL3 silencing in vivo and in vitro, we generated bcl3-deficient preclinical mouse models as well as murine cell lines and correlated our findings with human cell lines, PDX models, and 2 independent patient cohorts. We assessed the correlation of bcl3 expression pattern with clinical parameters and subtypes. RESULTS: Bcl3 was significantly down-regulated in human CSCs. Recapitulating this phenotype in preclinical mouse models of PDAC via BCL3 genetic knockout enhanced tumor burden, metastasis, epithelial to mesenchymal transition, and reduced overall survival. Fluorescence-activated cell sorting analyses, together with oxygen consumption, sphere formation, and tumorigenicity assays, all indicated that BCL3 loss resulted in CSC compartment expansion promoting cellular dedifferentiation. Overexpression of BCL3 in human PDXs diminished tumor growth by significantly reducing the CSC population and promoting differentiation. Human PDACs with low BCL3 expression correlated with increased metastasis, and BCL3-negative tumors correlated with lower survival and nonclassical subtypes. CONCLUSIONS: We demonstrate that bcl3 impacts pancreatic carcinogenesis by restraining CSC expansion and by curtailing an aggressive and metastatic tumor burden in PDAC across species. Levels of BCL3 expression are a useful stratification marker for predicting subtype characterization in PDAC, thereby allowing for personalized therapeutic approaches.


Subject(s)
B-Cell Lymphoma 3 Protein/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/metabolism , Animals , B-Cell Lymphoma 3 Protein/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/secondary , Cell Differentiation , Cell Line, Tumor , Cell Movement , Cell Proliferation , Energy Metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Signal Transduction , Tumor Burden , Tumor Cells, Cultured
8.
Sci Rep ; 11(1): 1191, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33441943

ABSTRACT

The in vivo assessment of tissue metabolism represents a novel strategy for the evaluation of oncologic disease. Hepatocellular carcinoma (HCC) is a high-prevalence, high-mortality tumor entity often discovered at a late stage. Recent evidence indicates that survival differences depend on metabolic alterations in tumor tissue, with particular focus on glucose metabolism and lactate production. Here, we present an in vivo imaging technique for metabolic tumor phenotyping in rat models of HCC. Endogenous HCC was induced in Wistar rats by oral diethyl-nitrosamine administration. Peak lactate-to-alanine signal ratios (L/A) were assessed with hyperpolarized magnetic resonance spectroscopic imaging (HPMRSI) after [1-13C]pyruvate injection. Cell lines were derived from a subset of primary tumors, re-implanted in nude rats, and assessed in vivo with dynamic hyperpolarized magnetic resonance spectroscopy (HPMRS) after [1-13C]pyruvate injection and kinetic modelling of pyruvate metabolism, taking into account systemic lactate production and recirculation. For ex vivo validation, enzyme activity and metabolite concentrations were spectroscopically quantified in cell and tumor tissue extracts. Mean peak L/A was higher in endogenous HCC compared to non-tumorous tissue. Dynamic HPMRS revealed higher pyruvate-to-lactate conversion rates (kpl) and lactate signal in subcutaneous tumors derived from high L/A tumor cells, consistent with ex vivo measurements of higher lactate dehydrogenase (LDH) levels in these cells. In conclusion, HPMRS and HPMRSI reveal distinct tumor phenotypes corresponding to differences in glycolytic metabolism in HCC tumor tissue.


Subject(s)
Carbon Isotopes/administration & dosage , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Magnetic Resonance Imaging/methods , Magnetic Resonance Spectroscopy/methods , Pyruvic Acid/administration & dosage , Alanine/metabolism , Animals , Cell Line, Tumor , Glycolysis/physiology , L-Lactate Dehydrogenase/metabolism , Lactic Acid/metabolism , Male , Rats , Rats, Nude , Rats, Wistar
9.
Acad Radiol ; 28 Suppl 1: S234-S243, 2021 11.
Article in English | MEDLINE | ID: mdl-33390324

ABSTRACT

RATIONALE AND OBJECTIVES: To investigate the effects of a reduced field-of-view (rFOV) acquisition in diffusion-weighted magnetic resonance imaging of the pancreas. MATERIALS AND METHODS: We enrolled 153 patients who underwent routine clinical MRI work-up including respiratory-triggered diffusion-weighted single-shot echo-planar imaging (DWI) with full field-of-view (fFOV, 3 × 3 × 4 mm3 voxel size) and reduced field-of-view (rFOV, 2.5 × 2.5 × 3 mm3 voxel size) for suspected pancreatic pathology. Two experienced radiologists were asked to subjectively rate (Likert Scale 1-4) image quality (overall image quality, lesion conspicuity, anatomical detail, artifacts). In addition, quantitative image parameters were assessed (apparent diffusion coefficient, apparent signal to noise ratio, apparent contrast to noise ratio [CNR]). RESULTS: All subjective metrics of image quality were rated in favor of rFOV DWI images compared to fFOV DWI images with substantial-to-high inter-rater reliability. Calculated ADC values of normal pancreas, pancreatic pathologies and reference tissues revealed no differences between both sequences. Whereas the apparent signal to noise ratio was higher in fFOV images, apparent CNR was higher in rFOV images. CONCLUSION: rFOV DWI provides higher image quality and apparent CNR values, favorable in the analysis of pancreatic pathologies.


Subject(s)
Diffusion Magnetic Resonance Imaging , Echo-Planar Imaging , Artifacts , Humans , Pancreas/diagnostic imaging , Reproducibility of Results
10.
Biomedicines ; 9(2)2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33513763

ABSTRACT

Hyperpolarized 13C nuclear magnetic resonance spectroscopy can characterize in vivo tissue metabolism, including preclinical models of cancer and inflammatory disease. Broad bandwidth radiofrequency excitation is often paired with free induction decay readout for spectral separation, but quantification of low-signal downstream metabolites using this method can be impeded by spectral peak overlap or when frequency separation of the detected peaks exceeds the excitation bandwidth. In this work, alternating frequency narrow bandwidth (250 Hz) slice-selective excitation was used for 13C spectroscopy at 7 T in a subcutaneous xenograft rat model of human pancreatic cancer (PSN1) to improve quantification while measuring the dynamics of injected hyperpolarized [1-13C]lactate and its metabolite [1-13C]pyruvate. This method does not require sophisticated pulse sequences or specialized radiofrequency and gradient pulses, but rather uses nominally spatially offset slices to produce alternating frequency excitation with simpler slice-selective radiofrequency pulses. Additionally, point-resolved spectroscopy was used to calibrate the 13C frequency from the thermal proton signal in the target region. This excitation scheme isolates the small [1-13C]pyruvate peak from the similar-magnitude tail of the much larger injected [1-13C]lactate peak, facilitates quantification of the [1-13C]pyruvate signal, simplifies data processing, and could be employed for other substrates and preclinical models.

11.
Nat Commun ; 11(1): 4527, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32913197

ABSTRACT

Evasion of programmed cell death represents a critical form of oncogene addiction in cancer cells. Understanding the molecular mechanisms underpinning cancer cell survival despite the oncogenic stress could provide a molecular basis for potential therapeutic interventions. Here we explore the role of pro-survival genes in cancer cell integrity during clonal evolution in non-small cell lung cancer (NSCLC). We identify gains of MCL-1 at high frequency in multiple independent NSCLC cohorts, occurring both clonally and subclonally. Clonal loss of functional TP53 is significantly associated with subclonal gains of MCL-1. In mice, tumour progression is delayed upon pharmacologic or genetic inhibition of MCL-1. These findings reveal that MCL-1 gains occur with high frequency in lung adenocarcinoma and can be targeted therapeutically.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Clonal Evolution , DNA Copy Number Variations , Datasets as Topic , Disease Models, Animal , Disease Progression , Humans , Lung/diagnostic imaging , Lung/pathology , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mice, Transgenic , Mutation , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Primary Cell Culture , Prospective Studies , Proto-Oncogene Proteins p21(ras)/genetics , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , RNA-Seq , Retrospective Studies , Spheroids, Cellular , Thiophenes/pharmacology , Thiophenes/therapeutic use , Tumor Burden/drug effects , Tumor Burden/genetics , Tumor Suppressor Protein p53/genetics , X-Ray Microtomography
12.
JCI Insight ; 5(15)2020 08 06.
Article in English | MEDLINE | ID: mdl-32614802

ABSTRACT

One of the major challenges in using pancreatic cancer patient-derived organoids (PDOs) in precision oncology is the time from biopsy to functional characterization. This is particularly true for endoscopic ultrasound-guided fine-needle aspiration biopsies, typically resulting in specimens with limited tumor cell yield. Here, we tested conditioned media of individual PDOs for cell-free DNA to detect driver mutations already early on during the expansion process to accelerate the genetic characterization of PDOs as well as subsequent functional testing. Importantly, genetic alterations detected in the PDO supernatant, collected as early as 72 hours after biopsy, recapitulate the mutational profile of the primary tumor, indicating suitability of this approach to subject PDOs to drug testing in a reduced time frame. In addition, we demonstrated that this workflow was practicable, even in patients for whom the amount of tumor material was not sufficient for molecular characterization by established means. Together, our findings demonstrate that generating PDOs from very limited biopsy material permits molecular profiling and drug testing. With our approach, this can be achieved in a rapid and feasible fashion with broad implications in clinical practice.


Subject(s)
Biomarkers, Tumor/genetics , Cell-Free Nucleic Acids/analysis , Cell-Free Nucleic Acids/genetics , Organoids/pathology , Pancreatic Neoplasms/pathology , Precision Medicine , Animals , Apoptosis , Biomarkers, Tumor/analysis , Cell Proliferation , Female , Humans , Mice , Mice, Nude , Organoids/metabolism , Pancreatic Neoplasms/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Eur J Radiol ; 124: 108848, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32006931

ABSTRACT

PURPOSE: To test combined dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and 18F-FDG positron emission tomography (FDG-PET)-derived parameters for prediction of histopathological grading in a rat Diethyl Nitrosamine (DEN)-induced hepatocellular carcinoma (HCC) model. METHODS: 15 male Wistar rats, aged 10 weeks were treated with oral DEN 0.01 % in drinking water and monitored until HCCs were detectable. DCE-MRI and PET were performed consecutively on small animal scanners. 38 tumors were identified and manually segmented based on HCC-specific contrast enhancement patterns. Grading (G2/3: 24 tumors, G1:14 tumors) alongside other histopathological parameters, tumor volume, contrast agent and 18F-FDG uptake metrics were noted. Class imbalance was addressed using SMOTE and collinearity was removed using hierarchical clustering and principal component analysis. A logistic regression model was fit separately to the individual parameter groups (DCE-MRI-derived, PET-derived, tumor volume) and the combined parameters. RESULTS: The combined model using all imaging-derived parameters achieved a mean ± STD sensitivity of 0.88 ± 0.16, specificity of 0.70 ± 0.20 and AUC of 0.90 ± 0.03. No correlation was found between tumor grading and tumor volume, morphology, necrosis, extracellular matrix, immune cell infiltration or underlying liver fibrosis. CONCLUSION: A combination of DCE-MRI- and 18F-FDG-PET-derived parameters provides high accuracy for histopathological grading of hepatocellular carcinoma in a relevant translational model system.


Subject(s)
Carcinoma, Hepatocellular/diagnostic imaging , Contrast Media , Fluorodeoxyglucose F18 , Image Enhancement/methods , Liver Neoplasms/diagnostic imaging , Magnetic Resonance Imaging/methods , Positron-Emission Tomography/methods , Animals , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Liver/diagnostic imaging , Liver/pathology , Liver Neoplasms/pathology , Male , Neoplasm Grading , Radiopharmaceuticals , Rats , Rats, Wistar , Sensitivity and Specificity , Tumor Burden
14.
EBioMedicine ; 15: 90-99, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28057438

ABSTRACT

Fluid-phase endocytosis is a homeostatic process with an unknown role in tumor initiation. The driver mutation in pancreatic ductal adenocarcinoma (PDAC) is constitutively active KRasG12D, which induces neoplastic transformation of acinar cells through acinar-to-ductal metaplasia (ADM). We have previously shown that KRasG12D-induced ADM is dependent on RAC1 and EGF receptor (EGFR) by a not fully clarified mechanism. Using three-dimensional mouse and human acinar tissue cultures and genetically engineered mouse models, we provide evidence that (i) KRasG12D leads to EGFR-dependent sustained fluid-phase endocytosis (FPE) during acinar metaplasia; (ii) variations in plasma membrane tension increase FPE and lead to ADM in vitro independently of EGFR; and (iii) that RAC1 regulates ADM formation partially through actin-dependent regulation of FPE. In addition, mice with a pancreas-specific deletion of the Neural-Wiskott-Aldrich syndrome protein (N-WASP), a regulator of F-actin, have reduced FPE and impaired ADM emphasizing the in vivo relevance of our findings. This work defines a new role of FPE as a tumor initiating mechanism.


Subject(s)
Endocytosis/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Precancerous Conditions , Proto-Oncogene Proteins p21(ras)/genetics , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Metaplasia , Mice , Mice, Knockout , Mutation , Osmotic Pressure , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics , Pancreatic Neoplasms
15.
Clin Cancer Res ; 23(6): 1461-1470, 2017 Mar 15.
Article in English | MEDLINE | ID: mdl-27663591

ABSTRACT

Purpose: Tumor heterogeneity is a hallmark of pancreatic ductal adenocarcinoma (PDAC). It determines tumor biology including tumor cellularity (i.e., amount of neoplastic cells and arrangement into clusters), which is related to the proliferative capacity and differentiation and the degree of desmoplasia among others. Given the close relation of tumor differentiation with differences in progression and therapy response or, e.g., the recently reported protective role of tumor stroma, we aimed at the noninvasive detection of PDAC groups, relevant for future personalized approaches. We hypothesized that histologic differences in PDAC tissue composition are detectable by the noninvasive diffusion weighted- (DW-) MRI-derived apparent diffusion coefficient (ADC) parameter.Experimental design: PDAC cellularity was quantified histologically and correlated with the ADC parameter and survival in genetically engineered mouse models and human patients.Results: Histologic analysis showed an inverse relationship of tumor cellularity and stroma content. Low tumor cellularity correlated with a significantly prolonged mean survival time (PDAClow = 21.93 months vs. PDACmed = 12.7 months; log-rank P < 0.001; HR = 2.23; CI, 1.41-3.53). Multivariate analysis using the Cox regression method confirmed tumor cellularity as an independent prognostic marker (P = 0.034; HR = 1.73; CI, 1.04-2.89). Tumor cellularity showed a strong negative correlation with the ADC parameter in murine (r = -0.84; CI, -0.90- -0.75) and human (r = -0.79; CI, -0.90 to -0.56) PDAC and high preoperative ADC values correlated with prolonged survival (ADChigh = 41.7 months; ADClow = 14.77 months; log rank, P = 0.040) in PDAC patients.Conclusions: This study identifies high tumor cellularity as a negative prognostic factor in PDAC and supports the ADC parameter for the noninvasive identification of PDAC groups. Clin Cancer Res; 23(6); 1461-70. ©2016 AACR.


Subject(s)
Adenocarcinoma/diagnostic imaging , Carcinoma, Pancreatic Ductal/diagnostic imaging , Diffusion Magnetic Resonance Imaging , Prognosis , Adenocarcinoma/diagnosis , Adenocarcinoma/pathology , Aged , Animals , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation , Disease Progression , Female , Humans , Male , Mice , Middle Aged
16.
J Nucl Med ; 57(12): 1971-1977, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27417649

ABSTRACT

Antibodies have become an established treatment modality in cancer therapy during the last decade. However, these treatments often suffer from an insufficient and heterogeneous response despite validated antigen or target receptor expression in the tumor. In fact, therapeutic success depends on both the presence of the tumor antigen and its accessibility by the antibody. In search of a suitable preclinical animal model to evaluate the mechanisms of tumor heterogeneity and hemodynamics, we characterized two exemplary non-Hodgkin lymphoma subtypes with comparable CD20 expression and metabolism, SUDHL-4 and Granta-519, using multimodal imaging techniques. METHODS: To investigate in vivo biodistribution, two differently modified αCD20 antigen-binding fragments (Fab), prepared by PASylation with a 200-residue polypeptide tag comprising Pro, Ala, and Ser (PAS200) and by fusion with an albumin-binding domain (ABD), were radiolabeled with 125I and intravenously injected into immunocompromised mice bearing corresponding xenografts. RESULTS: Validation with 18F-FDG revealed a similar distribution in vital tumor tissue 1 h after injection. However, large differences in tumor uptake were observed when the CD20-specific radiotracers 125I-Fab-ABD and 125I-Fab-PAS200 were applied (respective percentages injected dose per gram at 24 h after injection: 12.3 and 2.4 for Granta-519 vs. 5.8 and 1.2 for SUDHL-4). Three-dimensional light-sheet fluorescence microscopy with Cy5-Fab-PAS200 confirmed better tracer extravasation in the Granta-519 tumors. Moreover, dynamic contrast-enhanced (DCE) MRI revealed significantly reduced perfusion in the SUDHL-4 tumors. CONCLUSION: Tracer uptake was highly dependent on local tumor perfusion and Fab permeation in the SUDHL-4 and Granta-519 tumors. Thus, the SUDHL-4 xenograft offers an excellent model for investigating the influence of therapies affecting tumor angiogenesis.


Subject(s)
Antigens, CD20/immunology , Blood Circulation , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Humans , Mice , Microvessels/metabolism , Microvessels/physiopathology , Permeability , Protein Transport
17.
Oncotarget ; 7(31): 49156-49168, 2016 Aug 02.
Article in English | MEDLINE | ID: mdl-27203385

ABSTRACT

Surface CD24 has previously been described, together with CD44 and ESA, for the characterization of putative cancer stem cells in pancreatic ductal adenocarcinoma (PDAC), the most fatal of all solid tumors. CD24 has a variety of biological functions including the regulation of invasiveness and cell proliferation, depending on the tumor entity and subcellular localization. Genetically engineered mouse models (GEMM) expressing oncogenic KrasG12D recapitulate the human disease and develop PDAC. In this study we investigate the function of CD24 using GEMM of endogenous PDAC and a model of cerulein-induced acute pancreatitis. We found that (i) CD24 expression was upregulated in murine and human PDAC and during acute pancreatitis (ii) CD24 was expressed exclusively in differentiated PDAC, whereas CD24 absence was associated with undifferentiated tumors and (iii) membranous CD24 expression determines tumor subpopulations with an epithelial phenotype in grafted models. In addition, we show that CD24 protein is stabilized in response to WNT activation and that overexpression of CD24 in pancreatic cancer cells upregulated ß-catenin expression augmenting an epithelial, non-metastatic signature. Our results support a positive feedback model according to which (i) WNT activation and subsequent ß-catenin dephosphorylation stabilize CD24 protein expression, and (ii) sustained CD24 expression upregulates ß-catenin expression. Eventually, membranous CD24 augments the epithelial phenotype of pancreatic tumors. Thus we link the WNT/ß-catenin pathway with the regulation of CD24 in the context of PDAC differentiation.


Subject(s)
CD24 Antigen/metabolism , Cell Membrane/metabolism , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/metabolism , Animals , Carcinoma, Pancreatic Ductal/metabolism , Cell Differentiation , Cell Proliferation , Ceruletide/chemistry , Epithelial-Mesenchymal Transition , Epithelium/metabolism , Humans , Mice , Mice, Knockout , Mice, SCID , Neoplasm Transplantation , Pancreatitis/metabolism , Phenotype , Phosphorylation , Proto-Oncogene Proteins p21(ras)/genetics , Up-Regulation
18.
Nat Commun ; 7: 10770, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26916719

ABSTRACT

Mouse transgenesis has provided fundamental insights into pancreatic cancer, but is limited by the long duration of allele/model generation. Here we show transfection-based multiplexed delivery of CRISPR/Cas9 to the pancreas of adult mice, allowing simultaneous editing of multiple gene sets in individual cells. We use the method to induce pancreatic cancer and exploit CRISPR/Cas9 mutational signatures for phylogenetic tracking of metastatic disease. Our results demonstrate that CRISPR/Cas9-multiplexing enables key applications, such as combinatorial gene-network analysis, in vivo synthetic lethality screening and chromosome engineering. Negative-selection screening in the pancreas using multiplexed-CRISPR/Cas9 confirms the vulnerability of pancreatic cells to Brca2-inactivation in a Kras-mutant context. We also demonstrate modelling of chromosomal deletions and targeted somatic engineering of inter-chromosomal translocations, offering multifaceted opportunities to study complex structural variation, a hallmark of pancreatic cancer. The low-frequency mosaic pattern of transfection-based CRISPR/Cas9 delivery faithfully recapitulates the stochastic nature of human tumorigenesis, supporting wide applicability for biological/preclinical research.


Subject(s)
Carcinogenesis/genetics , Pancreas/metabolism , Pancreatic Neoplasms/genetics , Animals , BRCA2 Protein/genetics , CRISPR-Cas Systems , Chromosome Deletion , Electroporation , Genetic Engineering/methods , Genome , High-Throughput Nucleotide Sequencing , Immunohistochemistry , Magnetic Resonance Imaging , Mice , Mutation , Neoplasms, Experimental/genetics , Phylogeny , Polymerase Chain Reaction , Proto-Oncogene Proteins p21(ras)/genetics , Sequence Analysis, DNA , Transfection/methods , Translocation, Genetic/genetics
19.
Proc Natl Acad Sci U S A ; 112(45): 13982-7, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26508638

ABSTRACT

Here, we show CRISPR/Cas9-based targeted somatic multiplex-mutagenesis and its application for high-throughput analysis of gene function in mice. Using hepatic single guide RNA (sgRNA) delivery, we targeted large gene sets to induce hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). We observed Darwinian selection of target genes, which suppress tumorigenesis in the respective cellular/tissue context, such as Pten or Cdkn2a, and conversely found low frequency of Brca1/2 alterations, explaining mutational spectra in human ICC/HCC. Our studies show that multiplexed CRISPR/Cas9 can be used for recessive genetic screening or high-throughput cancer gene validation in mice. The analysis of CRISPR/Cas9-induced tumors provided support for a major role of chromatin modifiers in hepatobiliary tumorigenesis, including that of ARID family proteins, which have recently been reported to be mutated in ICC/HCC. We have also comprehensively characterized the frequency and size of chromosomal alterations induced by combinatorial sgRNA delivery and describe related limitations of CRISPR/Cas9 multiplexing, as well as opportunities for chromosome engineering in the context of hepatobiliary tumorigenesis. Our study describes novel approaches to model and study cancer in a high-throughput multiplexed format that will facilitate the functional annotation of cancer genomes.


Subject(s)
CRISPR-Cas Systems/genetics , Carcinoma, Hepatocellular/genetics , Disease Models, Animal , Genomics/methods , High-Throughput Screening Assays , Liver Neoplasms/genetics , Mutagenesis/genetics , Animals , Base Sequence , Gene Targeting , Histological Techniques , Liver/metabolism , Mice , Molecular Sequence Data , Selection, Genetic/genetics
20.
Nat Med ; 21(10): 1163-71, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26390243

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal human cancers and shows resistance to any therapeutic strategy used. Here we tested small-molecule inhibitors targeting chromatin regulators as possible therapeutic agents in PDAC. We show that JQ1, an inhibitor of the bromodomain and extraterminal (BET) family of proteins, suppresses PDAC development in mice by inhibiting both MYC activity and inflammatory signals. The histone deacetylase (HDAC) inhibitor SAHA synergizes with JQ1 to augment cell death and more potently suppress advanced PDAC. Finally, using a CRISPR-Cas9-based method for gene editing directly in the mouse adult pancreas, we show that de-repression of p57 (also known as KIP2 or CDKN1C) upon combined BET and HDAC inhibition is required for the induction of combination therapy-induced cell death in PDAC. SAHA is approved for human use, and molecules similar to JQ1 are being tested in clinical trials. Thus, these studies identify a promising epigenetic-based therapeutic strategy that may be rapidly implemented in fatal human tumors.


Subject(s)
Adenocarcinoma/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Epigenesis, Genetic , Histone Deacetylase Inhibitors/therapeutic use , Proteins/antagonists & inhibitors , Adenocarcinoma/therapy , Animals , Carcinoma, Pancreatic Ductal/therapy , Clustered Regularly Interspaced Short Palindromic Repeats , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...