Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Pathol ; 250(5): 685-692, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31994192

RESUMEN

Tissue diagnostics is the world of pathologists, and it is increasingly becoming digitalised to leverage the enormous potential of personalised medicine and of stratifying patients, enabling the administration of modern therapies. Therefore, the daily task for pathologists is changing drastically and will become increasingly demanding in order to take advantage of the development of modern computer technologies. The role of pathologist has rapidly evolved from exclusively describing the morphology and phenomenology of a disease, to becoming a gatekeeper for novel and most effective treatment options. This is possible based on the retrieval and management of a wide range of complex information from tissue or a group of cells and associated meta-data. Intelligent and self-learning software solutions can support and guide pathologists to score clinically relevant decisions based on the accurate and robust quantification of multiple target molecules or surrogate biomarker as companion or complimentary diagnostics along with relevant spatial relationships and contextual information from digital H&E and multiplexed images. With the availability of multiplex staining techniques on a single slide, high-resolution image analysis tools, and high-end computer hardware, machine and deep learning solutions now offer diagnostic rulesets and algorithms that still require clinical validation in well-designed studies. Before entering the clinical practice, the 'human factor' pathologist needs to develop trust in the output coming from the 'digital black box of computational pathology', including image analysis solutions and artificial intelligence algorithms to support critical clinical decisions which otherwise would not be available. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Algoritmos , Inteligencia Artificial , Interpretación de Imagen Asistida por Computador , Patólogos , Programas Informáticos , Humanos , Interpretación de Imagen Asistida por Computador/métodos , Patología/métodos
2.
Pathologe ; 42(5): 453-463, 2021 Sep.
Artículo en Alemán | MEDLINE | ID: mdl-34357472

RESUMEN

Most pancreatic ductal adenocarcinomas are localized in the pancreatic head. Due to the complex anatomic relationships with the surrounding organs and vascular structures in the retroperitoneal space and to the presence of numerous transection margins and dissection planes, pancreatic head resections belong to the most complex specimens concerning grossing and sampling for histopathologic analysis.Here we discuss current guidelines for standardized grossing and reporting of pancreatic cancer, with special reference to the assessment of the resection margin status. The importance of standardized reporting for the sake of completeness, comprehensibility, comparability, and quality control as well as for the integration of pathology reports in interdisciplinary digital workflows and artificial intelligence applications will be emphasized.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Inteligencia Artificial , Carcinoma Ductal Pancreático/diagnóstico , Humanos , Pancreatectomía , Neoplasias Pancreáticas/diagnóstico , Pancreaticoduodenectomía
3.
Mod Pathol ; 32(7): 1042-1052, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30737470

RESUMEN

Anti-angiogenic therapy and immune checkpoint inhibition are novel treatment strategies for patients with renal cell carcinoma. Various components and structures of the tumor microenvironment are potential predictive biomarkers and also attractive treatment targets. Macrophages, tumor infiltrating lymphocytes, vascular and lymphatic vessels represent an important part of the tumor immune environment, but their functional phenotypes and relevance for clinical outcome are yet ill defined. We applied Tissue Phenomics methods including image analysis for the standardized quantification of specific components and structures within the tumor microenvironment to profile tissue sections from 56 clear cell renal cell carcinoma patients. A characteristic composition and unique spatial relationship of CD68+ macrophages and tumor infiltrating lymphocytes correlated with overall survival. An inverse relationship was found between vascular (CD34) and lymphatic vessel (LYVE1) density. In addition, outcome was significantly better in patients with high blood vessel density in the tumors, whereas increased lymphatic vessel density in the tumors was associated with worse outcome. The Tissue Phenomics imaging analysis approach allowed visualization and simultaneous quantification of immune environment components, adding novel contextual information, and biological insights with potential applications in treatment response prediction.


Asunto(s)
Carcinoma de Células Renales/patología , Inmunohistoquímica/métodos , Neoplasias Renales/patología , Vasos Linfáticos/patología , Linfocitos Infiltrantes de Tumor/patología , Microambiente Tumoral/fisiología , Anciano , Carcinoma de Células Renales/metabolismo , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Neoplasias Renales/metabolismo , Vasos Linfáticos/metabolismo , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Persona de Mediana Edad
4.
Gastric Cancer ; 22(1): 77-90, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29779068

RESUMEN

BACKGROUND: Gastric cancer with lymphoid stroma (GCLS) is characterized by prominent stromal infiltration of T-lymphocytes. The aim of this study was to investigate GCLS biology through analysis of clinicopathological features, EBV infection, microsatellite instability (MSI), immune gene-expression profiling and PD-L1 status in neoplastic cells and tumor immune microenvironment. METHODS: Twenty-four GCLSs were analyzed by RNA in situ hybridization for EBV (EBER), PCR/fragment analysis for MSI, immunohistochemistry (PD-L1, cytokeratin, CD3, CD8), co-immunofluorescence (CK/PD-L1, CD68/PD-L1), NanoString gene-expression assay for immune-related genes and PD-L1 copy number alterations. CD3+ and CD8+ T-cell densities were calculated by digital analysis. Fifty-four non-GCLSs were used as control group. RESULTS: GCLSs displayed distinctive clinicopathological features, such as lower pTNM stage (p = 0.02) and better overall survival (p = 0.01). EBV+ or MSI-high phenotype was found in 66.7 and 16.7% cases, respectively. GCLSs harbored a cytotoxic T-cell-inflamed profile, particularly at the invasive front of tumors (p < 0.01) and in EBV+ cases (p = 0.01). EBV+ GCLSs, when compared to EBV- GCLSs, showed higher mRNA expression of genes related to Th1/cytotoxic and immunosuppressive biomarkers. PD-L1 protein expression, observed in neoplastic and immune stromal cells (33.3 and 91.7%, respectively), and PD-L1 amplification (18.8%) were restricted to EBV+/MSI-high tumors and correlated with high values of PD-L1 mRNA expression. CONCLUSIONS: This study shows that GCLS has a distinctive clinico-pathological and molecular profile. Furthermore, through an in-depth study of tumor immune microenvironment-by digital analysis and mRNA expression profiling-it highlights the role of EBV infection in promoting an inflamed tumor microenvironment, with putative therapeutic implications.


Asunto(s)
Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Gástricas/patología , Microambiente Tumoral/inmunología , Adulto , Anciano , Antígeno B7-H1/biosíntesis , Infecciones por Virus de Epstein-Barr/complicaciones , Femenino , Herpesvirus Humano 4 , Humanos , Inmunofenotipificación , Inflamación/genética , Inflamación/inmunología , Masculino , Inestabilidad de Microsatélites , Persona de Mediana Edad , Neoplasias Gástricas/genética , Neoplasias Gástricas/inmunología , Linfocitos T/inmunología , Linfocitos T/patología , Transcriptoma , Microambiente Tumoral/genética
5.
Chemistry ; 24(11): 2584-2587, 2018 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-29315897

RESUMEN

To date, many poly(ethylene glycol) (PEG) and poly(N-isopropylacrylamide) (PNIPAAm) biomolecule conjugates have been described, but they often show long response times, are not bio-inert, or lose function in biological fluids. Herein, we present a modular synthetic approach to generate polyvinylphosphonate biomolecule conjugates. These conjugates exhibit a sharp phase transition temperature even under physiological conditions where few other examples with this property have been described to date. Furthermore, it was feasible to add biological functions to the polymers via the conjugation step. The polyvinylphosphonate cholesterol constructs are attached to the cellular membrane and the folic acid anchored polymers are shuttled into the cells. This is an exceptional finding through a straightforward synthetic approach.


Asunto(s)
Colorantes Fluorescentes/química , Polivinilos/química , Línea Celular , Colorantes Fluorescentes/metabolismo , Ácido Fólico/química , Humanos , Microscopía Confocal , Ácidos Fosforosos/química , Polivinilos/metabolismo , Espectrofotometría
6.
BMC Cancer ; 15: 237, 2015 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-25879229

RESUMEN

BACKGROUND: Adenocarcinoma originating from the digestive system is a major contributor to cancer-related deaths worldwide. Tumor recurrence, advanced local growth and metastasis are key factors that frequently prevent these tumors from curative surgical treatment. Preclinical research has demonstrated that the dependency of these tumors on supporting mesenchymal stroma results in susceptibility to cell-based therapies targeting this stroma. METHODS/DESIGN: TREAT-ME1 is a prospective, uncontrolled, single-arm phase I/II study assessing the safety and efficacy of genetically modified autologous mesenchymal stromal cells (MSC) as delivery vehicles for a cell-based gene therapy for advanced, recurrent or metastatic gastrointestinal or hepatopancreatobiliary adenocarcinoma. Autologous bone marrow will be drawn from each eligible patient after consent for bone marrow donation has been obtained (under a separate EC-approved protocol). In the following ~10 weeks the investigational medicinal product (IMP) is developed for each patient. To this end, the patient's MSCs are stably transfected with a gamma-retroviral, replication-incompetent and self-inactivating (SIN) vector system containing a therapeutic promoter - gene construct that allows for tumor-specific expression of the therapeutic gene. After release of the IMP the patients are enrolled after given informed consent for participation in the TREAT-ME 1 trial. In the phase I part of the study, the safety of the IMP is tested in six patients by three treatment cycles consisting of re-transfusion of MSCs at different concentrations followed by administration of the prodrug Ganciclovir. In the phase II part of the study, sixteen patients will be enrolled receiving IMP treatment. A subgroup of patients that qualifies for surgery will be treated preoperatively with the IMP to verify homing of the MSCs to tumors as to be confirmed in the surgical specimen. DISCUSSION: The TREAT-ME1 clinical study involves a highly innovative therapeutic strategy combining cell and gene therapy and is conducted at a high level of pharmaceutical quality ensuring patient safety. This patient-tailored approach represents the first clinical study worldwide utilizing genetically engineered MSCs in humans. TRIAL REGISTRATION: EU Clinical Trials Register/European Union Drug Regulating Authorities Clinical Trials Database number: 2012-003741-15.


Asunto(s)
Protocolos Clínicos , Neoplasias Gastrointestinales/genética , Neoplasias Gastrointestinales/terapia , Terapia Genética , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Neoplasias Gastrointestinales/patología , Expresión Génica , Orden Génico , Genes Transgénicos Suicidas , Terapia Genética/efectos adversos , Vectores Genéticos/genética , Humanos , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia , Estadificación de Neoplasias
7.
Biotechnol Bioeng ; 111(11): 2290-302, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24890974

RESUMEN

Mesenchymal stromal cells (MSCs) are promising candidates for cell therapy. Their therapeutic use requires extensive expansion to obtain a sufficiently high number of cells for clinical applications. State-of-the-art expansion systems, that is, primarily culture flask-based systems, are limited regarding scale-up, automation, and reproducibility. To overcome this bottleneck, microcarrier (MC)-based expansion processes have been developed. For the first time, MSCs from the perinatal sources umbilical cord (UC) and amniotic membrane (AM) were expanded on MCs. This study focuses on the comparison of flask- and Cytodex 1 MC-expanded MSCs by evaluating the influence of the expansion process on biological MSC characteristics. Furthermore, we tested the hypothesis to obtain more homogeneous MSC preparations by expanding cells on MCs in controlled large-scale bioreactors. MSCs were extensively characterized determining morphology, cell growth, surface marker expression, and functional properties such as differentiation capacity, secretion of paracrine factors, and gene expression. Based on their gene expression profile MSCs from different donors and sources clearly clustered in distinct groups solely depending on the expansion process-MC or flask culture. MC- and flask-expanded MSCs significantly differed from each other regarding surface markers and both paracrine factors and gene expression profiles. Furthermore, based on gene expression analysis, MC cultivation of MSCs in controlled bioreactor systems resulted in less heterogeneity between cells from different donors. In conclusion, MC-based MSC expansion in controlled bioreactors has the potential to reliably produce MSCs with altered characteristics and functions as compared to flask-expanded MSCs. These findings may be useful for the generation of MSCs with tailored properties for clinical applications.


Asunto(s)
Reactores Biológicos , Células Madre Mesenquimatosas/fisiología , Técnicas de Cultivo de Célula/métodos , Proliferación Celular , Perfilación de la Expresión Génica , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo
8.
Cancer Rep (Hoboken) ; 7(1): e1916, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37950626

RESUMEN

BACKGROUND: Inflammatory myofibroblastic tumor (IMTs) are rare mesenchymal neoplasms with slow growth. Resection is considered as therapeutic standard, with chemotherapy being insufficiently effective in advanced disease. ALK translocations are present in 50% of cases, ROS1 fusions (YWHAE::ROS1, TFG::ROS1) are extremely rare. Here, we present a case with TFG::ROS1 fusion and highlight the significance of molecular tumor boards (MTBs) in clinical precision oncology for post-last-line therapy. CASE PRESENTATION: A 32-year-old woman presented with IMT diagnosed at age 27 for biopsy and treatment evaluation. Previous treatments included multiple resections and systemic therapy with vinblastine, cyclophosphamide, and methotrexate. A computed tomography scan showed extensive tumor infiltration of the psoas muscles and the posterior abdomen. Next generation sequencing revealed an actionable ROS1 fusion (TFG::ROS1) with breakpoints at exon 4/35 including the kinase domain and activating the RAS-pathway. TFG, the Trk-fused gene, exerts functions such as intracellular trafficking and exhibits high sequence homology between species. Based on single reports about efficacy of ROS1-targeting in ROS1 translocation positive IMTs the patient was started on crizotinib, an ATP-competitive small molecule c-MET, ALK and ROS1-inhibitor. With a follow-up of more than 9 months, the patient continues to show a profound response with major tumor regression, improved quality of life and no evidence for severe adverse events. CONCLUSION: This case underscores the importance of the availability of modern molecular diagnostics and interdisciplinarity in precision oncology to identify rare, disease-defining genotypes that make an otherwise difficult-to-treat disease targetable.


Asunto(s)
Neoplasias , Proteínas Tirosina Quinasas , Femenino , Humanos , Adulto , Proteínas Tirosina Quinasas/genética , Calidad de Vida , Proteínas Proto-Oncogénicas/genética , Medicina de Precisión , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas de Transporte Vesicular
9.
PLoS One ; 19(1): e0295641, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38215076

RESUMEN

Brain metastasis leads to increased mortality and is a major site of relapse for several cancers, yet the molecular mechanisms of brain metastasis are not well understood. In this study, we established and characterized a new leukemic cell line, FIA10, that metastasizes into the central nervous system (CNS) following injection into the tail vein of syngeneic mice. Mice injected with FIA10 cells developed neurological symptoms such as loss of balance, tremor, ataxic gait and seizures, leading to death within 3 months. Histopathology coupled with PCR analysis clearly showed infiltration of leukemic FIA10 cells into the brain parenchyma of diseased mice, with little involvement of bone marrow, peripheral blood and other organs. To define pathways that contribute to CNS metastasis, global transcriptome and proteome analysis was performed on FIA10 cells and compared with that of the parental stem cell line FDCP-Mix and the related FIA18 cells, which give rise to myeloid leukemia without CNS involvement. 188 expressed genes (RNA level) and 189 proteins were upregulated (log2 ratio FIA10/FIA18 ≥ 1) and 120 mRNAs and 177 proteins were downregulated (log2 ratio FIA10/FIA18 ≤ 1) in FIA10 cells compared with FIA18 cells. Major upregulated pathways in FIA10 cells revealed by biofunctional analyses involved immune response components, adhesion molecules and enzymes implicated in extracellular matrix remodeling, opening and crossing the blood-brain barrier (BBB), molecules supporting migration within the brain parenchyma, alterations in metabolism necessary for growth within the brain microenvironment, and regulators for these functions. Downregulated RNA and protein included several tumor suppressors and DNA repair enzymes. In line with the function of FIA10 cells to specifically infiltrate the brain, FIA10 cells have acquired a phenotype that permits crossing the BBB and adapting to the brain microenvironment thereby escaping immune surveillance. These data and our model system FIA10 will be valuable resources to study the occurrence of brain metastases and may help in the development of potential therapies against brain invasion.


Asunto(s)
Neoplasias Encefálicas , Neoplasias del Sistema Nervioso Central , Ratones , Animales , Transcriptoma , Proteómica , Encéfalo/metabolismo , Barrera Hematoencefálica/metabolismo , Neoplasias del Sistema Nervioso Central/patología , Neoplasias Encefálicas/patología , Perfilación de la Expresión Génica , ARN/metabolismo , Línea Celular , Microambiente Tumoral
10.
Cancer Rep (Hoboken) ; 6(7): e1796, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36813293

RESUMEN

BACKGROUND: The currently available immunotherapies already changed the strategy how many cancers are treated from first to last line. Understanding even the most complex heterogeneity in tumor tissue and mapping the spatial cartography of the tumor immunity allows the best and optimized selection of immune modulating agents to (re-)activate the patient's immune system and direct it against the individual cancer in the most effective way. RECENT FINDINGS: Primary cancer and metastases maintain a high degree of plasticity to escape any immune surveillance and continue to evolve depending on many intrinsic and extrinsic factors In the field of immune-oncology (IO) immune modulating agents are recognized as practice changing therapeutic modalities. Recent studies have shown that an optimal and lasting efficacy of IO therapeutics depends on the understanding of the spatial communication network and functional context of immune and cancer cells within the tumor microenvironment. Artificial intelligence (AI) provides an insight into the immune-cancer-network through the visualization of very complex tumor and immune interactions in cancer tissue specimens and allows the computer-assisted development and clinical validation of such digital biomarker. CONCLUSIONS: The successful implementation of AI-supported digital biomarker solutions guides the clinical selection of effective immune therapeutics based on the retrieval and visualization of spatial and contextual information from cancer tissue images and standardized data. As such, computational pathology (CP) turns into "precision pathology" delivering individual therapy response prediction. Precision Pathology does not only include digital and computational solutions but also high levels of standardized processes in the routine histopathology workflow and the use of mathematical tools to support clinical and diagnostic decisions as the basic principle of a "precision oncology".


Asunto(s)
Inteligencia Artificial , Neoplasias , Humanos , Neoplasias/terapia , Oncología Médica , Biomarcadores , Medicina de Precisión/métodos , Microambiente Tumoral
11.
Cancers (Basel) ; 14(20)2022 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-36291825

RESUMEN

(1) Background: molecular tumor boards (MTBs) are crucial instruments for discussing and allocating targeted therapies to suitable cancer patients based on genetic findings. Currently, limited evidence is available regarding the regional impact and the outreach component of MTBs; (2) Methods: we analyzed MTB patient data from four neighboring Bavarian tertiary care oncology centers in Würzburg, Erlangen, Regensburg, and Augsburg, together constituting the WERA Alliance. Absolute patient numbers and regional distribution across the WERA-wide catchment area were weighted with local population densities; (3) Results: the highest MTB patient numbers were found close to the four cancer centers. However, peaks in absolute patient numbers were also detected in more distant and rural areas. Moreover, weighting absolute numbers with local population density allowed for identifying so-called white spots-regions within our catchment that were relatively underrepresented in WERA MTBs; (4) Conclusions: investigating patient data from four neighboring cancer centers, we comprehensively assessed the regional impact of our MTBs. The results confirmed the success of existing collaborative structures with our regional partners. Additionally, our results help identifying potential white spots in providing precision oncology and help establishing a joint WERA-wide outreach strategy.

12.
Ann Surg ; 253(3): 566-71, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21169810

RESUMEN

OBJECTIVE: To specifically target tumor angiogenesis by linking transgene expression of engineered mesenchymal stem cells to angiopoietin-1-induced differentiation. BACKGROUND: Mesenchymal stem cells (MSCs) have been used to deliver therapeutic genes into solid tumors. These strategies rely on their homing mechanisms only to deliver the therapeutic agent. METHODS: We engineered murine MSC to express reporter genes or therapeutic genes under the selective control of the Tie2 promoter/enhancer. This approach uses the differentiative potential of MSCs induced by the tumor microenvironment to drive therapeutic gene expression only in the context of angiogenesis. RESULTS: When injected into the peripheral circulation of mice with either, orthotopic pancreatic or spontaneous breast cancer, the engineered MSCs were actively recruited to growing tumor vasculature and induced the selective expression of either reporter red florescent protein or suicide genes [herpes simplex virus-thymidine kinase (TK) gene] when the adoptively transferred MSC developed endothelial-like characteristics. The TK gene product in combination with the prodrug ganciclovir (GCV) produces a potent toxin, which affects replicative cells. The homing of engineered MSC with selective induction of TK in concert with GCV resulted in a toxic tumor-specific environment. The efficacy of this approach was demonstrated by significant reduction in primary tumor growth and prolongation of life in both tumor models. CONCLUSION: This "Trojan Horse" combined stem cell/gene therapy represents a novel treatment strategy for tailored therapy of solid tumors.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/patología , Angiopoyetina 1/farmacología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Expresión Génica/genética , Marcación de Gen , Genes Reporteros/genética , Genes Transgénicos Suicidas/genética , Ingeniería Genética , Terapia Genética/métodos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Neovascularización Patológica/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Timidina Quinasa/genética , Transgenes/genética , Animales , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Patológica/patología , Proteínas Tirosina Quinasas Receptoras/genética , Receptor TIE-2 , Simplexvirus/genética
13.
Ann Surg ; 254(5): 767-74; discussion 774-5, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22042469

RESUMEN

BACKGROUND: The use of engineered mesenchymal stem cells (MSCs) as therapeutic vehicles for the treatment of experimental pancreatic and breast cancer has been previously demonstrated. The potential application of MSCs for the treatment of hepatocellular carcinoma (HCC) has been controversial. The general approach uses engineered MSCs to target different aspects of tumor biology, including angiogenesis or the fibroblast-like stromal compartment, through the use of tissue-specific expression of therapeutic transgenes. The aim of the present study was (1) to evaluate the effect of exogenously added MSCs on the growth of HCC and (2) the establishment of an MSC-based suicide gene therapy for experimental HCC. METHODS: Mesenchymal stem cells were isolated from bone marrow of C57/Bl6 p53(-/-) mice. The cells were injected into mice with HCC xenografts and the effect on tumor proliferation and angiogenesis was evaluated. The cells were then stably transfected with red fluorescent protein (RFP) or Herpes simplex virus thymidine kinase (HSV-Tk) gene under control of the Tie2 promoter/enhancer or the CCL5 promoter. Mesenchymal stem cells were injected intravenously into mice with orthotopically growing xenografts of HCC and treated with ganciclovir (GCV). RESULTS: Ex vivo examination of hepatic tumors revealed tumor-specific recruitment, enhanced tumor growth, and increased microvessel density after nontherapeutic MSC injections. After their homing to the hepatic xenografts, engineered MSCs demonstrated activation of the Tie2 or CCL5 promoter as shown by RFP expression. Application of CCL5/HSV-TK transfected MSCs in combination with GCV significantly reduced tumor growth by 56.4% as compared with the control group and by 71.6% as compared with nontherapeutic MSC injections. CCL5/HSV-TK(+) transfected MSCs proved more potent in tumor inhibition as compared with Tie2/HSV-TK(+) MSCs. CONCLUSION: Exogenously added MSCs are recruited to growing HCC xenografts with concomitant activation of the CCL5 or Tie2 promoters within the MSCs. Stem cell-mediated introduction of suicide genes into the tumor followed by prodrug administration was effective for treatment of experimental HCC and thus may help fill the existing gap in bridging therapies for patients suffering from advanced HCCs.


Asunto(s)
Carcinoma Hepatocelular/terapia , Genes Transgénicos Suicidas/genética , Terapia Genética/métodos , Neoplasias Hepáticas Experimentales/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Microambiente Tumoral , Animales , Antivirales/uso terapéutico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/fisiopatología , Diferenciación Celular , Proliferación Celular , Quimiocina CCL5/genética , Modelos Animales de Enfermedad , Femenino , Ganciclovir/uso terapéutico , Ingeniería Genética , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/fisiopatología , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica , Proteínas Tirosina Quinasas Receptoras/genética , Receptor TIE-2 , Transfección , Trasplante Heterólogo
14.
Virchows Arch ; 478(2): 335-341, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32719890

RESUMEN

The rapid development of pathology is in contrast to a shortage of qualified staff. The aims of the present study are to compile basic information on the numbers of German physicians in pathology and to compare it with the situation in Europe and overseas. In addition, model calculations will shed light on the effects of part-time working models. Various publicly accessible databases (EuroStat) as well as publications of medical associations and professional associations of European countries and the USA/Canada were examined. In addition, a survey was carried out among the institutes of German universities. Figures from 24 European countries and the USA/Canada were evaluated. With one pathologist per 47,989 inhabitants, the density of pathologists in Germany in relation to the population is the second-lowest in Europe (average: 32,018). Moreover, the proportion of pathologists among the physicians working in Germany is the lowest in Europe and at the same time lower than in the USA and Canada (Germany: 1:200, USA: 1:70, Canada: 1:49). The ratio of pathologists to medical specialists is shifted in the same direction. The survey among university pathologists revealed a relevant increase in the workload over the last 10 years. The majority of institutes can manage this workload only with considerable difficulties. With a ratio between specialists and residents of 1:1, the university institutes show a high commitment in the area of training. The results of this study indicate a shortage of pathologists in Germany that could lead to a bottleneck in large parts of the health system.


Asunto(s)
Necesidades y Demandas de Servicios de Salud , Fuerza Laboral en Salud , Evaluación de Necesidades , Patólogos/provisión & distribución , Patología , Canadá , Selección de Profesión , Educación de Postgrado en Medicina , Alemania , Humanos , Patólogos/educación , Patología/educación , Especialización , Encuestas y Cuestionarios , Estados Unidos
15.
Cancers (Basel) ; 13(9)2021 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-33922988

RESUMEN

In this study, we developed the Binary ImaGe Colon Metastasis classifier (BIg-CoMet), a semi-guided approach for the stratification of colon cancer patients into two risk groups for the occurrence of distant metastasis, using an InceptionResNetV2-based deep learning model trained on binary images. We enrolled 291 colon cancer patients with pT3 and pT4 adenocarcinomas and converted one cytokeratin-stained representative tumor section per case into a binary image. Image augmentation and dropout layers were incorporated to avoid overfitting. In a validation collective (n = 128), BIg-CoMet was able to discriminate well between patients with and without metastasis (AUC: 0.842, 95% CI: 0.774-0.911). Further, the Kaplan-Meier curves of the metastasis-free survival showed a highly significant worse clinical course for the high-risk group (log-rank test: p < 0.001), and we demonstrated superiority over other established risk factors. A multivariable Cox regression analysis adjusted for confounders supported the use of risk groups as a prognostic factor for the occurrence of metastasis (hazard ratio (HR): 5.4, 95% CI: 2.5-11.7, p < 0.001). BIg-CoMet achieved good performance for both UICC subgroups, especially for UICC III (n = 53), with a positive predictive value of 80%. Our study demonstrates the ability to stratify colon cancer patients via a semi-guided process on images that primarily reflect tumor architecture.

16.
Lancet Digit Health ; 3(10): e654-e664, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34417147

RESUMEN

BACKGROUND: Response to immunotherapy in gastric cancer is associated with microsatellite instability (or mismatch repair deficiency) and Epstein-Barr virus (EBV) positivity. We therefore aimed to develop and validate deep learning-based classifiers to detect microsatellite instability and EBV status from routine histology slides. METHODS: In this retrospective, multicentre study, we collected tissue samples from ten cohorts of patients with gastric cancer from seven countries (South Korea, Switzerland, Japan, Italy, Germany, the UK and the USA). We trained a deep learning-based classifier to detect microsatellite instability and EBV positivity from digitised, haematoxylin and eosin stained resection slides without annotating tumour containing regions. The performance of the classifier was assessed by within-cohort cross-validation in all ten cohorts and by external validation, for which we split the cohorts into a five-cohort training dataset and a five-cohort test dataset. We measured the area under the receiver operating curve (AUROC) for detection of microsatellite instability and EBV status. Microsatellite instability and EBV status were determined to be detectable if the lower bound of the 95% CI for the AUROC was above 0·5. FINDINGS: Across the ten cohorts, our analysis included 2823 patients with known microsatellite instability status and 2685 patients with known EBV status. In the within-cohort cross-validation, the deep learning-based classifier could detect microsatellite instability status in nine of ten cohorts, with AUROCs ranging from 0·597 (95% CI 0·522-0·737) to 0·836 (0·795-0·880) and EBV status in five of eight cohorts, with AUROCs ranging from 0·819 (0·752-0·841) to 0·897 (0·513-0·966). Training a classifier on the pooled training dataset and testing it on the five remaining cohorts resulted in high classification performance with AUROCs ranging from 0·723 (95% CI 0·676-0·794) to 0·863 (0·747-0·969) for detection of microsatellite instability and from 0·672 (0·403-0·989) to 0·859 (0·823-0·919) for detection of EBV status. INTERPRETATION: Classifiers became increasingly robust when trained on pooled cohorts. After prospective validation, this deep learning-based tissue classification system could be used as an inexpensive predictive biomarker for immunotherapy in gastric cancer. FUNDING: German Cancer Aid and German Federal Ministry of Health.


Asunto(s)
Aprendizaje Profundo , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/diagnóstico , Inestabilidad de Microsatélites , Neoplasias Gástricas/complicaciones , Neoplasias Gástricas/genética , Anciano , Estudios de Cohortes , Femenino , Alemania , Técnicas Histológicas/métodos , Humanos , Italia , Japón , Masculino , Persona de Mediana Edad , Reproducibilidad de los Resultados , República de Corea , Estudios Retrospectivos , Suiza , Reino Unido , Estados Unidos
17.
Circulation ; 119(2): 281-9, 2009 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-19118255

RESUMEN

BACKGROUND: The importance and therapeutic value of stem cells in lymphangiogenesis are poorly understood. We evaluated the potential of human and murine mesenchymal stem cells (MSCs) to acquire a lymphatic phenotype in vitro and to enhance lymphatic regeneration in vivo. METHODS AND RESULTS: We assessed the lymphendothelial differentiation of human and murine MSCs after induction with supernatant derived from human dermal microvascular endothelial cells, isolated lymphatic endothelial cells, and purified vascular endothelial growth factor (VEGF)-C in vitro. We used human or murine progenitor MSC lines and then characterized the lymphatic phenotype by morphology, migratory capacity, and the expression of lymphatic markers such as Prox-1, podoplanin, Lyve-1, VEGF receptor-2, and VEGF receptor-3. Using a murine lymphatic edema model, we assessed the potential of these cells to form a functional lymphatic vasculature in vivo after injection of syngeneic MSCs. Incubation with supernatant from lymphatic endothelial cells induced an endothelium-like morphology and the expression of lymphendothelial markers in both human and murine MSCs in vitro. MSCs showed migratory activity along a VEGF-C gradient, which was enhanced by VEGF-C conditioning. In vivo, the local application of MSCs resulted in a significant decrease in edema formation (-20.1%; P<0.01 versus untreated tails) after 3 weekly cell injections and restored the drainage of intradermally injected methylene blue after 7 weekly injections. CONCLUSIONS: MSCs were capable of expressing a lymphatic phenotype when exposed to lymph-inductive media and purified VEGF-C. Migratory activity toward VEGF-C in vitro suggests homing capability in vivo. Restoration of lymphatic drainage after injection of MSCs in a lymphedema model indicates that MSCs play a role in lymphatic regeneration. The potential clinical application of MSC in wound healing and reduction of lymphatic edema warrants further research.


Asunto(s)
Endotelio Linfático/fisiología , Linfangiogénesis/fisiología , Vasos Linfáticos/fisiología , Células Madre Mesenquimatosas/fisiología , Células Madre Multipotentes/fisiología , Fenotipo , Regeneración/fisiología , Animales , Diferenciación Celular/fisiología , Línea Celular , Movimiento Celular/fisiología , Endotelio Linfático/citología , Femenino , Humanos , Vasos Linfáticos/citología , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células Madre Multipotentes/citología
18.
Cytotherapy ; 12(7): 853-6, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20942603

RESUMEN

The last decade has seen a dramatic rise in the development of new cellular therapeutics in a wide range of indications. There have been acceptable safety profiles reported in early studies using blood-derived and adherent stem cell products, but also an inconsistent efficacy record. Further expansion has been hindered in part by a lack of capital (both private and public) and delayed entry into the cell therapy space by large healthcare and pharmaceutical companies, those members of the industry most reliably able to initiate and maintain advanced-phase clinical trials. With recognition that the International Society for Cellular Therapy (ISCT) is uniquely positioned to serve the global translational regenerative medicine research community as a network hub for scientific standards and policy, the ISCT commissioned the establishment of an Industry Task Force (ITF) to address current and future roles for industry. The objectives of the ITF were to gather information and prioritize efforts for a new Commercialization Committee (CC) and to construct innovative platforms that would foster constructive and synergistic collaborations between industry and ISCT. Recommendations and conclusions of the ITF included that the new CC: (1) foster new relationships with therapeutic and stem cell societies, (2) foster educational workshops and forums to cross-educate and standardize practices, (3) create industry subcommittees to address priority initiatives, with clear benchmarks and global implementation, and (4) establish a framework for a greater industry community within ISCT, opening doors for industry to share the new vision for commercialization of cell therapy, emphasizing the regenerative medicine space.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Ensayos Clínicos como Asunto , Industria Farmacéutica , Comercio , Humanos , Guías de Práctica Clínica como Asunto , Medicina Regenerativa , Sociedades Científicas , Investigación Biomédica Traslacional
19.
J Pathol Clin Res ; 6(4): 273-282, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32592447

RESUMEN

The biological complexity reflected in histology images requires advanced approaches for unbiased prognostication. Machine learning and particularly deep learning methods are increasingly applied in the field of digital pathology. In this study, we propose new ways to predict risk for cancer-specific death from digital images of immunohistochemically (IHC) stained tissue microarrays (TMAs). Specifically, we evaluated a cohort of 248 gastric cancer patients using convolutional neural networks (CNNs) in an end-to-end weakly supervised scheme independent of subjective pathologist input. To account for the time-to-event characteristic of the outcome data, we developed new survival models to guide the network training. In addition to the standard H&E staining, we investigated the prognostic value of a panel of immune cell markers (CD8, CD20, CD68) and a proliferation marker (Ki67). Our CNN-derived risk scores provided additional prognostic value when compared to the gold standard prognostic tool TNM stage. The CNN-derived risk scores were also shown to be superior when systematically compared to cell density measurements or a CNN score derived from binary 5-year survival classification, which ignores time-to-event. To better understand the underlying biological mechanisms, we qualitatively investigated risk heat maps for each marker which visualised the network output. We identified patterns of biological interest that were related to low risk of cancer-specific death such as the presence of B-cell predominated clusters and Ki67 positive sub-regions and showed that the corresponding risk scores had prognostic value in multivariate Cox regression analyses (Ki67&CD20 risks: hazard ratio (HR) = 1.47, 95% confidence interval (CI) = 1.15-1.89, p = 0.002; CD20&CD68 risks: HR = 1.33, 95% CI = 1.07-1.67, p = 0.009). Our study demonstrates the potential additional value that deep learning in combination with a panel of IHC markers can bring to the field of precision oncology.


Asunto(s)
Biomarcadores de Tumor/análisis , Aprendizaje Profundo , Interpretación de Imagen Asistida por Computador , Inmunohistoquímica , Neoplasias Gástricas/química , Microambiente Tumoral , Antígenos CD/análisis , Antígenos CD20/análisis , Antígenos de Diferenciación Mielomonocítica/análisis , Antígenos CD8/análisis , Proliferación Celular , Humanos , Antígeno Ki-67/análisis , Estadificación de Neoplasias , Valor Predictivo de las Pruebas , Medición de Riesgo , Factores de Riesgo , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Factores de Tiempo , Análisis de Matrices Tisulares
20.
Ann Surg ; 250(5): 747-53, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19826249

RESUMEN

OBJECTIVE: To analyze the efficacy of engineered mesenchymal stem cell based therapy directed towards pancreatic tumor stroma. SUMMARY BACKGROUND DATA: Mesenchymal stem cells (MSC) are actively recruited to tumor stroma where they enhance tumor growth and metastases. Upregulation of chemotactic cytokine (CCL5) by MSCs within the tumor stroma has been shown to play a central role in this process. Murine MSCs were engineered to express reporter genes or therapeutic genes under control of the CCL5 promoter and adoptively transferred into mice with growing pancreatic tumors. The effect on tumor growth and metastases was then evaluated. METHODS: MSCs isolated from bone marrow of C57/Bl6 p53 mice were stably transfected with red fluorescent protein (RFP), enhanced green fluorescent protein (eGFP), or herpes simplex virus (HSV) thymidine kinase (Tk) gene driven by the RANTES promoter. MSCs were intravenously applied once per week over 3 weeks to mice carrying an orthotopic, syngeneic pancreatic Panc02 tumor. RESULTS: eGFP and RFP signals driven by the CCL5 promoter were detected by fluorescence in treated pancreatic tumor samples. The HSV-Tk therapy group treated intraperitoneal with the prodrug ganciclovir 5 to 7 days after stem cell application lead to a 50% reduction of primary pancreatic tumor growth (P < 0.0003, student t test) and reduced liver metastases (0% vs. 60%). CONCLUSION: The active homing of MSCs into primary pancreatic tumor stroma and activation of the CCL5 promoter was verified using eGFP- and RFP-reporter genes. In the presence of ganciclovir, HSV-Tk transfected MSCs led to a significant reduction of primary pancreatic tumor growth and incidence of metastases.


Asunto(s)
Genes Transgénicos Suicidas , Terapia Genética , Células Madre Mesenquimatosas/metabolismo , Neoplasias Pancreáticas/terapia , Ingeniería de Tejidos , Animales , Movimiento Celular , Quimiocina CCL5/genética , Genes Reporteros , Inyecciones Intraperitoneales , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/patología , Regiones Promotoras Genéticas , Simplexvirus/genética , Timidina Quinasa/genética , Timidina Quinasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA