Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
MAGMA ; 37(2): 257-272, 2024 Apr.
Article En | MEDLINE | ID: mdl-38366129

OBJECTIVE: To compensate subject-specific field inhomogeneities and enhance fat pre-saturation with a fast online individual spectral-spatial (SPSP) single-channel pulse design. METHODS: The RF shape is calculated online using subject-specific field maps and a predefined excitation k-space trajectory. Calculation acceleration options are explored to increase clinical viability. Four optimization configurations are compared to a standard Gaussian spectral selective pre-saturation pulse and to a Dixon acquisition using phantom and volunteer (N = 5) data at 1.5 T with a turbo spin echo (TSE) sequence. Measurements and simulations are conducted across various body parts and image orientations. RESULTS: Phantom measurements demonstrate up to a 3.5-fold reduction in residual fat signal compared to Gaussian fat saturation. In vivo evaluations show improvements up to sixfold for dorsal subcutaneous fat in sagittal cervical spine acquisitions. The versatility of the tailored trajectory is confirmed through sagittal foot/ankle, coronal, and transversal cervical spine experiments. Additional measurements indicate that excitation field (B1) information can be disregarded at 1.5 T. Acceleration methods reduce computation time to a few seconds. DISCUSSION: An individual pulse design that primarily compensates for main field (B0) inhomogeneities in fat pre-saturation is successfully implemented within an online "push-button" workflow. Both fat saturation homogeneity and the level of suppression are improved.


Image Enhancement , Magnetic Resonance Imaging , Humans , Magnetic Resonance Imaging/methods , Image Enhancement/methods , Imaging, Three-Dimensional/methods , Phantoms, Imaging , Heart Rate , Cervical Vertebrae/diagnostic imaging
2.
Magn Reson Med ; 89(1): 77-94, 2023 Jan.
Article En | MEDLINE | ID: mdl-36128895

PURPOSE: To evaluate the benefits and challenges of dynamic parallel transmit (pTx) pulses for fat saturation (FS) and water-excitation (WE), in the context of CEST MRI. METHODS: "Universal" kT -points (for FS) and spiral non-selective (for WE) trajectories were optimized offline for flip angle (FA) homogeneity. Routines to optimize the pulse shape online, based on the subject's fields maps, were implemented (target FA of 110°/0° for FS, 0°/5° for WE at fat/water frequencies). The pulses were inserted in a CEST sequence with a pTx readout. The different fat suppression schemes and their effects on CEST contrasts were compared in 12 volunteers at 7T. RESULTS: With a 25%-shorter pulse duration, pTx FS largely improved the FA homogeneity (root-mean-square-error (RMSE) = 12.3° vs. 53.4° with circularly-polarized mode, at the fat frequency). However, the spectral selectivity was degraded mainly in the cerebellum and close to the sinuses (RMSE = 5.8° vs. 0.2° at the water frequency). Similarly, pTx WE showed a trade-off between FA homogeneity and spectral selectivity compared to pTx non-selective pulses (RMSE = 0.9° and 1.1° at the fat and water frequencies, vs. 4.6° and 0.5°). In the brain, CEST metrics were reduced by up to 31.9% at -3.3 ppm with pTx FS, suggesting a mitigated lipid-induced bias. CONCLUSION: This clinically compatible implementation of dynamic pTx pulses improved the fat suppression homogeneity at 7T taking into account the subject-specific B0 heterogeneities online. This study highlights the lipid-induced biases on the CEST z-spectrum. The results are promising for body applications where B0 heterogeneities and fat are more substantial.


Magnetic Resonance Imaging , Water , Humans , Magnetic Resonance Imaging/methods , Brain/diagnostic imaging , Contrast Media , Lipids , Algorithms
3.
Magn Reson Med ; 87(1): 207-219, 2022 01.
Article En | MEDLINE | ID: mdl-34411335

PURPOSE: To improve pseudo-continuous arterial spin labeling (pcASL) at 7T by exploiting a hybrid homogeneity- and efficiency-optimized B1+ -shim with adapted gradient strength as well as background suppression. METHODS: The following three experiments were performed at 7T, each employing five volunteers: (1) A hybrid (ie, homogeneity-efficiency optimized) B1+ -shim was introduced and evaluated for variable-rate selective excitation pcASL labeling. Therefore, B1+ -maps in the V3 segment and time-of-flight images were acquired to identify the feeding arteries. For validation, a gradient-echo sequence was applied in circular polarized (CP) mode and with the hybrid B1+ -shim. Additionally, the gray matter (temporal) signal-to-noise ratio (tSNR) in pcASL perfusion images was evaluated. (2) Bloch simulations for the pcASL labeling were conducted and validated experimentally, with a focus on the slice-selective gradients. (3) Background suppression was added to the B1+ -shimmed, gradient-adapted 7T sequence and this was then compared to a matched sequence at 3T. RESULTS: The B1+ -shim improved the signal within the labeling plane (23.6%) and the SNR/tSNR increased (+11%/+11%) compared to its value in CP mode; however, the increase was not significant. In accordance with the simulations, the adapted gradients increased the tSNR (35%) and SNR (45%) significantly. Background suppression further improved the perfusion images at 7T, and this protocol performed as well as a resolution-matched protocol at 3T. CONCLUSION: The combination of the proposed hybrid B1+ -phase-shim with the adapted slice-selective gradients and background suppression shows great potential for improved pcASL labeling under suboptimal B1+ conditions at 7T.


Arteries , Brain , Brain/diagnostic imaging , Cerebrovascular Circulation , Gray Matter/diagnostic imaging , Signal-To-Noise Ratio , Spin Labels
4.
Mol Cancer Res ; 19(4): 688-701, 2021 04.
Article En | MEDLINE | ID: mdl-33443114

Specific biological properties of those circulating cancer cells that are the origin of brain metastases (BM) are not well understood. Here, single circulating breast cancer cells were fate-tracked during all steps of the brain metastatic cascade in mice after intracardial injection over weeks. A novel in vivo two-photon microscopy methodology was developed that allowed to determine the specific cellular and molecular features of breast cancer cells that homed in the brain, extravasated, and successfully established a brain macrometastasis. Those BM-initiating breast cancer cells (BMIC) were mainly originating from a slow-cycling subpopulation that included only 16% to 20% of all circulating cancer cells. BMICs showed enrichment of various markers of cellular stemness. As a proof of principle for the principal usefulness of this approach, expression profiling of BMICs versus non-BMICs was performed, which revealed upregulation of NDRG1 in the slow-cycling BMIC subpopulation in one BM model. Here, BM development was completely suppressed when NDRG1 expression was downregulated. In accordance, in primary human breast cancer, NDRG1 expression was heterogeneous, and high NDRG1 expression was associated with shorter metastasis-free survival. In conclusion, our data identify temporary slow-cycling breast cancer cells as the dominant source of brain and other metastases and demonstrates that this can lead to better understanding of BMIC-relevant pathways, including potential new approaches to prevent BM in patients. IMPLICATIONS: Cancer cells responsible for successful brain metastasis outgrowth are slow cycling and harbor stemness features. The molecular characteristics of these metastasis-initiating cells can be studied using intravital microscopy technology.


Brain Neoplasms/secondary , Brain/physiopathology , Neoplastic Cells, Circulating/metabolism , Animals , Cell Line, Tumor , Female , Humans , Mice , Neoplasm Metastasis
5.
Pancreas ; 46(3): 311-322, 2017 03.
Article En | MEDLINE | ID: mdl-27846146

OBJECTIVES: Pancreatic ductal adenocarcinoma (PDAC) has been subclassified into 3 molecular subtypes: classical, quasi-mesenchymal, and exocrine-like. These subtypes exhibit differences in patient survival and drug resistance to conventional therapies. The aim of the current study is to identify novel subtype-specific protein biomarkers facilitating subtype stratification of patients with PDAC and novel therapy development. METHODS: A set of 12 human patient-derived primary cell lines was used as a starting material for an advanced label-free proteomics approach leading to the identification of novel cell surface and secreted biomarkers. Cell surface protein identification was achieved by in vitro biotinylation, followed by mass spectrometric analysis of purified biotin-tagged proteins. Proteins secreted into a chemically defined serum-free cell culture medium were analyzed by shotgun proteomics. RESULTS: Of 3288 identified proteins, 2 pan-PDAC (protocadherin-1 and lipocalin-2) and 2 exocrine-like-specific (cadherin-17 and galectin-4) biomarker candidates have been validated. Proximity ligation assay analysis of the 2 exocrine-like biomarkers revealed their co-localization on the surface of exocrine-like cells. CONCLUSIONS: The study reports the identification and validation of novel PDAC biomarkers relevant for the development of patient stratification tools. In addition, cadherin-17 and galectin-4 may serve as targets for bispecific antibodies as novel therapeutics in PDAC.


Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms/metabolism , Proteome/metabolism , Proteomics/methods , Animals , Cadherins/metabolism , Carcinoma, Pancreatic Ductal/diagnosis , Galectin 4/metabolism , Humans , Lipocalin-2/metabolism , Mice , Pancreatic Neoplasms/diagnosis , Protocadherins , Reproducibility of Results , Sensitivity and Specificity , Transplantation, Heterologous
6.
J Cell Mol Med ; 20(4): 721-30, 2016 Apr.
Article En | MEDLINE | ID: mdl-26849468

The long-term propagation of basal prostate progenitor cells ex vivo has been very difficult in the past. The development of novel methods to expand prostate progenitor cells in vitro allows determining their cell surface phenotype in greater detail. Mouse (Lin(-)Sca-1(+) CD49f(+) Trop2(high)-phenotype) and human (Lin(-) CD49f(+) TROP2(high)) basal prostate progenitor cells were expanded in vitro. Human and mouse cells were screened using 242 anti-human or 176 antimouse monoclonal antibodies recognizing the cell surface protein profile. Quantitative expression was evaluated at the single-cell level using flow cytometry. Differentially expressed cell surface proteins were evaluated in conjunction with the known CD49f(+)/TROP2(high) phenotype of basal prostate progenitor cells and characterized by in vivo sandwich-transplantation experiments using nude mice. The phenotype of basal prostate progenitor cells was determined as CD9(+)/CD24(+)/CD29(+)/CD44(+)/CD47(+)/CD49f(+)/CD104(+)/CD147(+)/CD326(+)/Trop2(high) of mouse as well as human origin. Our analysis revealed several proteins, such as CD13, Syndecan-1 and stage-specific embryonal antigens (SSEAs), as being differentially expressed on murine and human CD49f(+) TROP2(+) basal prostate progenitor cells. Transplantation experiments suggest that CD49f(+) TROP2(high) SSEA-4(high) human prostate basal progenitor cells to be more potent to regenerate prostate tubules in vivo as compared with CD49f(+) TROP2(high) or CD49f(+) TROP2(high) SSEA-4(low) cells. Determination of the cell surface protein profile of functionally defined murine and human basal prostate progenitor cells reveals differentially expressed proteins that may change the potency and regenerative function of epithelial progenitor cells within the prostate. SSEA-4 is a candidate cell surface marker that putatively enables a more accurate identification of the basal PESC lineage.


Epithelial Cells/transplantation , Regeneration/genetics , Stage-Specific Embryonic Antigens/genetics , Stem Cell Transplantation , Stem Cells/cytology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Proliferation , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Male , Mice , Mice, Nude , Prostate/surgery , Signal Transduction , Stage-Specific Embryonic Antigens/metabolism , Stem Cells/metabolism , Syndecan-1/genetics , Syndecan-1/metabolism
7.
Nat Med ; 22(3): 278-87, 2016 Mar.
Article En | MEDLINE | ID: mdl-26855150

Although subtypes of pancreatic ductal adenocarcinoma (PDAC) have been described, this malignancy is clinically still treated as a single disease. Here we present patient-derived models representing the full spectrum of previously identified quasi-mesenchymal (QM-PDA), classical and exocrine-like PDAC subtypes, and identify two markers--HNF1A and KRT81--that enable stratification of tumors into different subtypes by using immunohistochemistry. Individuals with tumors of these subtypes showed substantial differences in overall survival, and their tumors differed in drug sensitivity, with the exocrine-like subtype being resistant to tyrosine kinase inhibitors and paclitaxel. Cytochrome P450 3A5 (CYP3A5) metabolizes these compounds in tumors of the exocrine-like subtype, and pharmacological or short hairpin RNA (shRNA)-mediated CYP3A5 inhibition sensitizes tumor cells to these drugs. Whereas hepatocyte nuclear factor 4, alpha (HNF4A) controls basal expression of CYP3A5, drug-induced CYP3A5 upregulation is mediated by the nuclear receptor NR1I2. CYP3A5 also contributes to acquired drug resistance in QM-PDA and classical PDAC, and it is highly expressed in several additional malignancies. These findings designate CYP3A5 as a predictor of therapy response and as a tumor cell-autonomous detoxification mechanism that must be overcome to prevent drug resistance.


Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/genetics , Cytochrome P-450 CYP3A/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 1-alpha/metabolism , Keratins, Hair-Specific/metabolism , Keratins, Type II/metabolism , Pancreatic Neoplasms/genetics , Aged , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Dasatinib/therapeutic use , Erlotinib Hydrochloride/therapeutic use , Female , Hepatocyte Nuclear Factor 4/metabolism , Humans , Immunohistochemistry , Male , Mice, Inbred NOD , Middle Aged , Neoplasm Transplantation , Paclitaxel/therapeutic use , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pregnane X Receptor , Prognosis , Protein Kinase Inhibitors/therapeutic use , Receptors, Steroid/metabolism , Up-Regulation
8.
Prostate ; 76(1): 114-21, 2016 Jan.
Article En | MEDLINE | ID: mdl-26444457

BACKGROUND: Based on findings of surface marker, protein screens as well as the postulated near-urethral location of the prostate stem cell niche, we were interested whether androgen ablation, distinct anatomic regions within the prostate or neurotrophins have an influence on basal prostate epithelial progenitor cells (PESCs). METHODS: Microdissection of the prostate, enzymatic digestion, and preparation of single cells was performed from murine and human prostates. Adult PESC marker expressions were compared between a group of C57BL/6 mice and a separate group of castrated C57BL/6 mice. Surface markers CD13/CD271 on human prostate epithelial progenitor cells were evaluated by FACS analyses in cells cultured under novel stem cell conditions. The effect of neurotrophins NGF, NT3, and BDNF were evaluated with respect to their influence on proliferation and activation of human basal PESCs in vitro. RESULTS: We demonstrate the highest percentage of CD49f+ and Trop2+ expressing cells in the urethra near prostatic regions of WT mice (Trop2+ proximal: 10% vs. distal to the urethra: 3%, P < 0.001). While a marked increase of Trop2 expressing cells can be measured both in the proximal and distal prostatic regions after castration, the most prominent increase in Trop2+ cells can be measured in the prostatic tissue distant to the urethra. Furthermore, we demonstrate that the proportion of syndecan-1 expressing cells greatly increases in the regions proximal to the urethra after castration (WT: 5% vs. castrated: 40%). We identified heterogeneous CD13 and nerve growth factor receptor (p75(NGFR), CD271) expression on CD49f(+)/TROP2(high) human basal PESCs. Addition of the neurotrophins NT3, BDNF, and NGF to the stem cell media led to a marked temporary increase in the proliferation of human basal PESCs. CONCLUSIONS: Our results in mice support the model, in which the proximal urethral region contains the prostate stem cell niche while a stronger androgen-dependent regulation of adult prostate stem cells can be found in the peripheral prostatic tissue. Neutrophin signaling via nerve growth factor receptor is possibly involved in human prostate stem cell homeostasis.


Antigens, Neoplasm/metabolism , Epithelial Cells , Nerve Growth Factors/metabolism , Prostate , Prostatic Neoplasms , Urethra , Animals , Antigens, CD/metabolism , Castration/methods , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Urethra/metabolism , Urethra/pathology
9.
Stem Cell Reports ; 4(3): 503-18, 2015 Mar 10.
Article En | MEDLINE | ID: mdl-25702639

Methods to isolate and culture primary prostate epithelial stem/progenitor cells (PESCs) have proven difficult and ineffective. Here, we present a method to grow and expand both murine and human basal PESCs long term in serum- and feeder-free conditions. The method enriches for adherent mouse basal PESCs with a Lin(-)SCA-1(+)CD49f(+)TROP2(high) phenotype. Progesterone and sodium selenite are additionally required for the growth of human Lin(-)CD49f(+)TROP2(high) PESCs. The gene-expression profiles of expanded basal PESCs show similarities to ESCs, and NF-kB function is critical for epithelial differentiation of sphere-cultured PESCs. When transplanted in combination with urogenital sinus mesenchyme, expanded mouse and human PESCs generate ectopic prostatic tubules, demonstrating their stem cell activity in vivo. This novel method will facilitate the molecular, genomic, and functional characterization of normal and pathologic prostate glands of mouse and human origin.


Cell Culture Techniques , Cell Separation , Prostate/cytology , Stem Cells/cytology , Animals , Cell Adhesion , Cell Differentiation , Cell Self Renewal , Cell Separation/methods , Culture Media, Serum-Free , Gene Expression Profiling , Humans , Immunomagnetic Separation/methods , Male , Mice , NF-kappa B/metabolism , Progesterone/metabolism , Progesterone/pharmacology , Prostate/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Sodium Selenite/pharmacology , Stem Cells/drug effects , Stem Cells/metabolism , Transcriptome , Tumor Necrosis Factor-alpha/metabolism
10.
Cell Stem Cell ; 14(6): 824-37, 2014 Jun 05.
Article En | MEDLINE | ID: mdl-24704494

Myelodysplastic syndromes (MDSs) are a heterogeneous group of myeloid neoplasms with defects in hematopoietic stem and progenitor cells (HSPCs) and possibly the HSPC niche. Here, we show that patient-derived mesenchymal stromal cells (MDS MSCs) display a disturbed differentiation program and are essential for the propagation of MDS-initiating Lin(-)CD34(+)CD38(-) stem cells in orthotopic xenografts. Overproduction of niche factors such as CDH2 (N-Cadherin), IGFBP2, VEGFA, and LIF is associated with the ability of MDS MSCs to enhance MDS expansion. These factors represent putative therapeutic targets in order to disrupt critical hematopoietic-stromal interactions in MDS. Finally, healthy MSCs adopt MDS MSC-like molecular features when exposed to hematopoietic MDS cells, indicative of an instructive remodeling of the microenvironment. Therefore, this patient-derived xenograft model provides functional and molecular evidence that MDS is a complex disease that involves both the hematopoietic and stromal compartments. The resulting deregulated expression of niche factors may well also be a feature of other hematopoietic malignancies.


Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Stem Cell Niche , Aged , Animals , Humans , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID
11.
Urol Oncol ; 32(5): 678-86, 2014 Jul.
Article En | MEDLINE | ID: mdl-24631171

OBJECTIVES: To evaluate CD24/CD44/CD47 cancer stem cell marker expressions in bladder cancer (BCa) and provide data on their prognostic significance for clinical outcome in patients undergoing radical cystectomy (RC). MATERIAL AND METHODS: Primary BCa tissue was used for xenograft studies. A tissue microarray was prepared using specimens from a cohort of 132 patients. All patients underwent RC for urothelial BCa between 2001 and 2010. Expression of CD24, CD44, and CD47 was examined in primary samples and xenografts by fluorescence-activated cell sorting. Populations of CD24(low)- and CD24(high)-expressing cells were sorted and evaluated for tumorigenicity in vivo. Tissue microarray was analyzed for CD24/CD44 staining intensity and tumor-specific vs. stromal cell staining. Associations with BCa survival, BCa stage, and lymph node status were evaluated by univariate and multivariate analyses. RESULTS: CD24 and CD44/CD47 expressions mark distinct cell populations within the normal urothelium as well as in BCa. CD24(high/low) expression was not sufficient to characterize CD24 as a BCa-initiating marker in in vivo primary xenotransplants. CD24 and CD44 expressions correlated with lower cancer-specific survival in patients. However, multivariate analyses of CD24 or CD44 did not demonstrate significantly increased hazards for cancer-specific death if analyzed together with stage, grade, and nodal status of patients. CONCLUSIONS: Cancer stem cell markers CD24/CD44/CD47 are differentially expressed in cells of urothelial BCa in patients undergoing RC and influence cancer-specific survival of patients. Further evaluation of CD24/CD44/CD47 protein expression could be of high therapeutic value in BCa. However, both CD24 and CD44 expressions cannot be regarded as independent prognostic parameters for patients undergoing RC.


CD24 Antigen/metabolism , CD47 Antigen/metabolism , Hyaluronan Receptors/metabolism , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/surgery , Urothelium/metabolism , Aged , Animals , Biomarkers, Tumor/metabolism , Cohort Studies , Cystectomy , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis , Male , Mice , Middle Aged , Multivariate Analysis , Neoplasm Transplantation , Prognosis , Tissue Array Analysis , Treatment Outcome , Urinary Bladder Neoplasms/diagnosis , Urothelium/pathology
12.
World J Urol ; 32(6): 1537-42, 2014 Dec.
Article En | MEDLINE | ID: mdl-24370691

PURPOSE: To evaluate the influence of type 2 diabetes on cancer-specific outcome in patients undergoing surgery for localized renal cell carcinoma (RCC). METHODS: A total of 1,140 patients with localized RCC undergoing radical or partial nephrectomy were enrolled into this retrospective case-control study. Primary outcome was the cancer-specific survival comparing patients with and without type 2 diabetes at the time of surgery. Secondary outcomes were recurrence-free survival and metastases-free survival comparing the same groups. Additionally, the influence of accompanying factors on cancer-specific survival and overall survival of patients was evaluated in a multivariate analysis. Among 1,140 patients included in the analyses, 202 had diabetes at the time of surgery and 938 patients without diabetes served as control. RESULTS: The univariate comparisons between patients with and without diabetes regarding recurrence-free, metastases-free, and cancer-specific survival revealed no significant differences. Multivariate results demonstrate that age, BMI, and diabetes had no significant effect on cancer-specific hazard among participants. After adjustment of the factors in terms of overall survival, however, increased age, increased BMI, and type 2 diabetes at the time of surgery were independent risk factors for the occurrence of the event death. CONCLUSIONS: Type 2 diabetes and obesity at the time of surgery have no significant impact on cancer-specific and recurrence-free survival in patients with localized renal cancer.


Carcinoma, Renal Cell/complications , Carcinoma, Renal Cell/mortality , Diabetes Mellitus, Type 2/complications , Kidney Neoplasms/complications , Kidney Neoplasms/mortality , Aged , Body Mass Index , Carcinoma, Renal Cell/surgery , Diabetes Mellitus, Type 2/mortality , Diabetes Mellitus, Type 2/pathology , Disease-Free Survival , Female , Humans , Kidney Neoplasms/surgery , Male , Middle Aged , Nephrectomy , Retrospective Studies , Risk Factors , Survival Rate , Treatment Outcome
13.
J Urol ; 190(6): 2263-70, 2013 Dec.
Article En | MEDLINE | ID: mdl-23820058

PURPOSE: For rare cancers such as neuroendocrine bladder cancer treatment options are limited due partly to the lack of preclinical models. Techniques to amplify rare primary neuroendocrine bladder cancer cells could provide novel tools for the discovery of drug and diagnostic targets. We developed preclinical experimental models for neuroendocrine bladder cancer. MATERIALS AND METHODS: Fresh tumor tissue from 2 patients with neuroendocrine bladder cancer was used to establish in vitro and in vivo models. We analyzed additional archived tissues in the National Center of Tumor Diseases tissue bank from patients with neuroendocrine bladder cancer. Primary tumor samples were collected during radical cystectomy. PHA-665752 was used to inhibit MET in animal models and cell cultures. The expression of markers and drug targets in neuroendocrine bladder cancer was determined by flow cytometry. The growth of neuroendocrine bladder cancer in vitro was determined by counting live cells. Tumor growth in mice was assessed by measuring tumor volume. Groups were compared using the nonparametric Kruskal-Wallis test. RESULTS: Xenograft models and serum-free cultures of neuroendocrine bladder cancer cells allowed screening for cell surface markers and drug targets. We found expression of the HGF receptor MET in neuroendocrine bladder cancer cultures, xenograft models and primary patient sections. The growth of neuroendocrine bladder cancer spheroids in vitro depended critically on HGF. Treatment of neuroendocrine bladder cancer bearing mice with a MET inhibitor significantly decreased tumor growth compared to that in control treated mice. CONCLUSIONS: Neuroendocrine bladder cancer xenografts and serum-free cultures provided suitable models in which to identify diagnostic markers and therapeutic targets. Using the models, we noted HGF dependent growth of human neuroendocrine bladder cancer and identified MET as a new treatment target for neuroendocrine bladder cancer.


Disease Models, Animal , Neuroendocrine Tumors , Urinary Bladder Neoplasms , Animals , Biomedical Research/methods , Heterografts , Humans , Mice , Neoplasm Transplantation , Proto-Oncogene Mas , Tumor Cells, Cultured
14.
J Immunol ; 189(12): 5602-11, 2012 Dec 15.
Article En | MEDLINE | ID: mdl-23152559

Myeloid-derived suppressor cells (MDSCs) represent a heterogeneous population of myeloid cells in cancer patients and tumor-bearing mice that potently inhibits T cell responses. During tumor progression, MDSCs accumulate in several organs, including the tumor tissue. So far, tumor-infiltrating MDSC subpopulations remain poorly explored. In this study, we performed global gene expression profiling of mouse tumor-infiltrating granulocytic and monocytic (MO-MDSC) subsets compared with MDSCs from peripheral blood. RMA-S lymphoma-infiltrating MO-MDSCs not only produced high levels of NO and arginase-1, but also greatly increased levels of chemokines comprising the CCR5 ligands CCL3, CCL4, and CCL5. MO-MDSCs isolated from B16 melanoma and from skin tumor-bearing ret transgenic mice also expressed high levels of CCL3, CCL4, and CCL5. Expression of CCR5 was preferentially detected on regulatory T cells (Tregs). Accordingly, tumor-infiltrating MO-MDSCs directly attracted high numbers of Tregs via CCR5 in vitro. Intratumoral injection of CCL4 or CCL5 increased tumor-infiltrating Tregs, and deficiency of CCR5 led to their profound decrease. Moreover, in CCR5-deficient mice, RMA-S and B16 tumor growth was delayed emphasizing the importance of CCR5 in the control of antitumor immune responses. Overall, our data demonstrate that chemokines secreted by tumor-infiltrating MO-MDSCs recruit high numbers of Tregs revealing a novel suppressive role of MDSCs with potential clinical implications for the development of cancer immunotherapies.


Chemotaxis, Leukocyte/immunology , Immune Tolerance/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Monocytes/immunology , Myeloid Cells/immunology , Receptors, CCR5/physiology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line, Tumor , Chemokine CCL3/biosynthesis , Chemokine CCL3/genetics , Chemokine CCL3/metabolism , Chemokine CCL4/biosynthesis , Chemokine CCL4/genetics , Chemokine CCL4/metabolism , Chemokine CCL5/biosynthesis , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Chemotaxis, Leukocyte/genetics , Disease Models, Animal , Immune Tolerance/genetics , Ligands , Lymphoma/immunology , Lymphoma/metabolism , Lymphoma/pathology , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Monocytes/metabolism , Monocytes/pathology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Receptors, CCR5/deficiency , Receptors, CCR5/metabolism , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology
15.
PLoS One ; 7(11): e48503, 2012.
Article En | MEDLINE | ID: mdl-23152778

Standard cancer cell lines do not model the intratumoural heterogeneity situation sufficiently. Clonal selection leads to a homogeneous population of cells by genetic drift. Heterogeneity of tumour cells, however, is particularly critical for therapeutically relevant studies, since it is a prerequisite for acquiring drug resistance and reoccurrence of tumours. Here, we report the isolation of a highly tumourigenic primary pancreatic cancer cell line, called JoPaca-1 and its detailed characterization at multiple levels. Implantation of as few as 100 JoPaca-1 cells into immunodeficient mice gave rise to tumours that were histologically very similar to the primary tumour. The high heterogeneity of JoPaca-1 was reflected by diverse cell morphology and a substantial number of chromosomal aberrations. Comparative whole-genome sequencing of JoPaca-1 and BxPC-3 revealed mutations in genes frequently altered in pancreatic cancer. Exceptionally high expression of cancer stem cell markers and a high clonogenic potential in vitro and in vivo was observed. All of these attributes make this cell line an extremely valuable model to study the biology of and pharmaceutical effects on pancreatic cancer.


Cell Transformation, Neoplastic , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , AC133 Antigen , Aldehyde Dehydrogenase 1 Family , Alleles , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antimetabolites, Antineoplastic/pharmacology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Genomic Instability , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Keratins/genetics , Keratins/metabolism , Male , Mesothelin , Mice , Middle Aged , Mutation , Neoplasm Metastasis , Pancreatic Neoplasms/metabolism , Peptides/genetics , Peptides/metabolism , Polyploidy , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism , Transplantation, Heterologous , Tumor Microenvironment , Gemcitabine
16.
Stem Cells ; 29(3): 418-29, 2011 Mar.
Article En | MEDLINE | ID: mdl-21308861

The identification of novel approaches to specifically target the DNA-damage checkpoint response in chemotherapy-resistant cancer stem cells (CSC) of solid tumors has recently attracted great interest. We show here in colon cancer cell lines and primary colon cancer cells that inhibition of checkpoint-modulating phosphoinositide 3-kinase-related (PIK) kinases preferentially depletes the chemoresistant and exclusively tumorigenic CD133(+) cell fraction. We observed a time- and dose-dependent disproportionally pronounced loss of CD133(+) cells and the consecutive lack of in vitro and in vivo tumorigenicity of the remaining cells. Depletion of CD133(+) cells was initiated through apoptosis of cycling CD133(+) cells and further substantiated through subsequent recruitment of quiescent CD133(+) cells into the cell cycle followed by their elimination. Models using specific PIK kinase inhibitors, somatic cell gene targeting, and RNA interference demonstrated that the observed detrimental effects of caffeine on CSC were attributable specifically to the inhibition of the PIK kinase ataxia telangiectasia- and Rad3-related (ATR). Mechanistically, phosphorylation of CHK1 checkpoint homolog (S. pombe; CHK1) was significantly enhanced in CD133(+) as compared with CD133(-) cells on treatment with DNA interstrand-crosslinking (ICL) agents, indicating a preferential activation of the ATR/CHK1-dependent DNA-damage response in tumorigenic CD133(+) cells. Consistently, the chemoresistance of CD133(+) cells toward DNA ICL agents was overcome through inhibition of ATR/CHK1-signaling. In conclusion, our study illustrates a novel target to eliminate the tumorigenic CD133(+) cell population in colon cancer and provides another rationale for the development of specific ATR-inhibitors.


Carcinoma/pathology , Cell Cycle Proteins/antagonists & inhibitors , Cell Transformation, Neoplastic/genetics , Colonic Neoplasms/pathology , Neoplastic Stem Cells/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , AC133 Antigen , Animals , Antigens, CD/metabolism , Ataxia Telangiectasia Mutated Proteins , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/therapy , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Cell Line, Tumor , Cell Separation/methods , Cell Transformation, Neoplastic/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/therapy , Down-Regulation/drug effects , Down-Regulation/genetics , Down-Regulation/physiology , Female , Gene Expression Regulation, Neoplastic/drug effects , Genetic Therapy , Glycoproteins/metabolism , Humans , Mice , Mice, Nude , Molecular Targeted Therapy/methods , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Peptides/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , RNA, Small Interfering/pharmacology , Xenograft Model Antitumor Assays
...