Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Cells ; 12(23)2023 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-38067116

RESUMEN

Many solid tumors are characterized by a dense extracellular matrix (ECM) composed of various ECM fibril proteins. These proteins provide structural support and a biological context for the residing cells. The reciprocal interactions between growing and migrating tumor cells and the surrounding stroma result in dynamic changes in the ECM architecture and its properties. With the use of advanced imaging techniques, several specific patterns in the collagen surrounding the breast tumor have been identified in both tumor murine models and clinical histology images. These tumor-associated collagen signatures (TACS) include loosely organized fibrils far from the tumor and fibrils aligned either parallel or perpendicular to tumor colonies. They are correlated with tumor behavior, such as benign growth or invasive migration. However, it is not fully understood how one specific fibril pattern can be dynamically remodeled to form another alignment. Here, we present a novel multi-cellular lattice-free (MultiCell-LF) agent-based model of ECM that, in contrast to static histology images, can simulate dynamic changes between TACSs. This model allowed us to identify the rules of cell-ECM physical interplay and feedback that guided the emergence and transition among various TACSs.


Asunto(s)
Colágeno , Neoplasias , Animales , Ratones , Colágeno/metabolismo , Colágenos Fibrilares/metabolismo , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Neoplasias/metabolismo
2.
BMC Biol ; 20(1): 163, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35840963

RESUMEN

INTRODUCTION: Aggressive cancers commonly ferment glucose to lactic acid at high rates, even in the presence of oxygen. This is known as aerobic glycolysis, or the "Warburg Effect." It is widely assumed that this is a consequence of the upregulation of glycolytic enzymes. Oncogenic drivers can increase the expression of most proteins in the glycolytic pathway, including the terminal step of exporting H+ equivalents from the cytoplasm. Proton exporters maintain an alkaline cytoplasmic pH, which can enhance all glycolytic enzyme activities, even in the absence of oncogene-related expression changes. Based on this observation, we hypothesized that increased uptake and fermentative metabolism of glucose could be driven by the expulsion of H+ equivalents from the cell. RESULTS: To test this hypothesis, we stably transfected lowly glycolytic MCF-7, U2-OS, and glycolytic HEK293 cells to express proton-exporting systems: either PMA1 (plasma membrane ATPase 1, a yeast H+-ATPase) or CA-IX (carbonic anhydrase 9). The expression of either exporter in vitro enhanced aerobic glycolysis as measured by glucose consumption, lactate production, and extracellular acidification rate. This resulted in an increased intracellular pH, and metabolomic analyses indicated that this was associated with an increased flux of all glycolytic enzymes upstream of pyruvate kinase. These cells also demonstrated increased migratory and invasive phenotypes in vitro, and these were recapitulated in vivo by more aggressive behavior, whereby the acid-producing cells formed higher-grade tumors with higher rates of metastases. Neutralizing tumor acidity with oral buffers reduced the metastatic burden. CONCLUSIONS: Therefore, cancer cells which increase export of H+ equivalents subsequently increase intracellular alkalization, even without oncogenic driver mutations, and this is sufficient to alter cancer metabolism towards an upregulation of aerobic glycolysis, a Warburg phenotype. Overall, we have shown that the traditional understanding of cancer cells favoring glycolysis and the subsequent extracellular acidification is not always linear. Cells which can, independent of metabolism, acidify through proton exporter activity can sufficiently drive their metabolism towards glycolysis providing an important fitness advantage for survival.


Asunto(s)
Neoplasias , Protones , Glucosa/metabolismo , Glucólisis/fisiología , Células HEK293 , Humanos , Ácido Láctico/metabolismo , Neoplasias/metabolismo
3.
Cancer Res ; 77(9): 2242-2254, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28249898

RESUMEN

Ongoing intratumoral evolution is apparent in molecular variations among cancer cells from different regions of the same tumor, but genetic data alone provide little insight into environmental selection forces and cellular phenotypic adaptations that govern the underlying Darwinian dynamics. In three spontaneous murine cancers (prostate cancers in TRAMP and PTEN mice, pancreatic cancer in KPC mice), we identified two subpopulations with distinct niche construction adaptive strategies that remained stable in culture: (i) invasive cells that produce an acidic environment via upregulated aerobic glycolysis; and (ii) noninvasive cells that were angiogenic and metabolically near-normal. Darwinian interactions of these subpopulations were investigated in TRAMP prostate cancers. Computer simulations demonstrated invasive, acid-producing (C2) cells maintain a fitness advantage over noninvasive, angiogenic (C3) cells by promoting invasion and reducing efficacy of immune response. Immunohistochemical analysis of untreated tumors confirmed that C2 cells were invariably more abundant than C3 cells. However, the C2 adaptive strategy phenotype incurred a significant cost due to inefficient energy production (i.e., aerobic glycolysis) and depletion of resources for adaptations to an acidic environment. Mathematical model simulations predicted that small perturbations of the microenvironmental extracellular pH (pHe) could invert the cost/benefit ratio of the C2 strategy and select for C3 cells. In vivo, 200 mmol/L NaHCO3 added to the drinking water of 4-week-old TRAMP mice increased the intraprostatic pHe by 0.2 units and promoted proliferation of noninvasive C3 cells, which remained confined within the ducts so that primary cancer did not develop. A 0.2 pHe increase in established tumors increased the fraction of C3 cells and signficantly diminished growth of primary and metastatic tumors. In an experimental tumor construct, MCF7 and MDA-MB-231 breast cancer cells were coinjected into the mammary fat pad of SCID mice. C2-like MDA-MB-231 cells dominated in untreated animals, but C3-like MCF7 cells were selected and tumor growth slowed when intratumoral pHe was increased. Overall, our data support the use of mathematical modeling of intratumoral Darwinian interactions of environmental selection forces and cancer cell adaptive strategies. These models allow the tumor to be steered into a less invasive pathway through the application of small but selective biological force. Cancer Res; 77(9); 2242-54. ©2017 AACR.


Asunto(s)
Neoplasias de la Mama/genética , Evolución Molecular , Neoplasias Pancreáticas/genética , Neoplasias de la Próstata/genética , Selección Genética/genética , Animales , Neoplasias de la Mama/patología , Linaje de la Célula/genética , Proliferación Celular/genética , Simulación por Computador , Femenino , Humanos , Células MCF-7 , Masculino , Ratones , Modelos Teóricos , Fosfohidrolasa PTEN/genética , Neoplasias Pancreáticas/patología , Neoplasias de la Próstata/patología , Miembro 25 de Receptores de Factores de Necrosis Tumoral/genética , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Theor Biol ; 414: 254-268, 2017 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-27890575

RESUMEN

We develop an off-lattice, agent-based model to describe vasculogenesis, the de novo formation of blood vessels from endothelial progenitor cells during development. The endothelial cells that comprise our vessel network are viewed as linearly elastic spheres that move in response to the forces they experience. We distinguish two types of endothelial cells: vessel elements are contained within the network and tip cells are located at the ends of vessels. Tip cells move in response to mechanical forces caused by interactions with neighbouring vessel elements and the local tissue environment, chemotactic forces and a persistence force which accounts for their tendency to continue moving in the same direction. Vessel elements are subject to similar mechanical forces but are insensitive to chemotaxis. An angular persistence force representing interactions with the local tissue is introduced to stabilise buckling instabilities caused by cell proliferation. Only vessel elements proliferate, at rates which depend on their degree of stretch: elongated elements have increased rates of proliferation, and compressed elements have reduced rates. Following division, the fate of the new cell depends on the local mechanical environment: the probability of forming a new sprout is increased if the parent vessel is highly compressed and the probability of being incorporated into the parent vessel increased if the parent is stretched. Simulation results reveal that our hybrid model can reproduce the key qualitative features of vasculogenesis. Extensive parameter sensitivity analyses show that significant changes in network size and morphology are induced by varying the chemotactic sensitivity of tip cells, and the sensitivities of the proliferation rate and the sprouting probability to mechanical stretch. Varying the chemotactic sensitivity directly influences the directionality of the networks. The degree of branching, and thereby the density of the networks, is influenced by the sprouting probability. Glyphs that simultaneously depict several network properties are introduced to show how these and other network quantities change over time and also as model parameters vary. We also show how equivalent glyphs constructed from in vivo data could be used to discriminate between normal and tumour vasculature and, in the longer term, for model validation. We conclude that our biomechanical hybrid model can generate vascular networks that are qualitatively similar to those generated from in vitro and in vivo experiments.


Asunto(s)
División Celular , Quimiotaxis , Células Endoteliales , Modelos Cardiovasculares , Neoplasias , Neovascularización Patológica , Animales , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Neoplasias/química , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Ratas
5.
Adv Exp Med Biol ; 936: 1-10, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27739040

RESUMEN

In the field of pathology it is clear that molecular genomics and digital imaging represent two promising future directions, and both are as relevant to the tumor microenvironment as they are to the tumor itself (Beck AH et al. Sci Transl Med 3(108):108ra113-08ra113, 2011). Digital imaging, or whole slide imaging (WSI), of glass histology slides facilitates a number of value-added competencies which were not previously possible with the traditional analog review of these slides under a microscope by a pathologist. As an important tool for investigational research, digital pathology can leverage the quantification and reproducibility offered by image analysis to add value to the pathology field. This chapter will focus on the application of image analysis to investigate the tumor microenvironment and how quantitative investigation can provide deeper insight into our understanding of the tumor to tumor microenvironment relationship.


Asunto(s)
Interpretación de Imagen Asistida por Computador/métodos , Procesamiento de Imagen Asistido por Computador/estadística & datos numéricos , Neoplasias/diagnóstico , Coloración y Etiquetado/métodos , Microambiente Tumoral , Eosina Amarillenta-(YS) , Colorantes Fluorescentes , Hematoxilina , Técnicas Histológicas , Humanos , Microscopía Fluorescente/instrumentación , Microscopía Fluorescente/métodos , Neoplasias/patología , Neoplasias/ultraestructura
6.
Cancer Res ; 76(11): 3136-44, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27009166

RESUMEN

Spatial heterogeneity in tumors is generally thought to result from branching clonal evolution driven by random mutations that accumulate during tumor development. However, this concept rests on the implicit assumption that cancer cells never evolve to a fitness maximum because they can always acquire mutations that increase proliferative capacity. In this study, we investigated the validity of this assumption. Using evolutionary game theory, we demonstrate that local cancer cell populations will rapidly converge to the fittest phenotype given a stable environment. In such settings, cellular spatial heterogeneity in a tumor will be largely governed by regional variations in environmental conditions, for example, alterations in blood flow. Model simulations specifically predict a common spatial pattern in which cancer cells at the tumor-host interface exhibit invasion-promoting, rapidly proliferating phenotypic properties, whereas cells in the tumor core maximize their population density by promoting supportive tissue infrastructures, for example, to promote angiogenesis. We tested model predictions through detailed quantitative image analysis of phenotypic spatial distribution in histologic sections of 10 patients with stage 2 invasive breast cancers. CAIX, GLUT1, and Ki67 were upregulated in the tumor edge, consistent with an acid-producing invasive, proliferative phenotype. Cells in the tumor core were 20% denser than the edge, exhibiting upregulation of CAXII, HIF-1α, and cleaved caspase-3, consistent with a more static and less proliferative phenotype. Similarly, vascularity was consistently lower in the tumor center compared with the tumor edges. Lymphocytic immune responses to tumor antigens also trended to higher level in the tumor edge, although this effect did not reach statistical significance. Like invasive species in nature, cancer cells at the leading edge of the tumor possess a different phenotype from cells in the tumor core. Our results suggest that at least some of the molecular heterogeneity in cancer cells in tumors is governed by predictable regional variations in environmental selection forces, arguing against the assumption that cancer cells can evolve toward a local fitness maximum by random accumulation of mutations. Cancer Res; 76(11); 3136-44. ©2016 AACR.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/patología , Modelos Teóricos , Mutación/genética , Microambiente Tumoral , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Anhidrasa Carbónica IX/genética , Anhidrasa Carbónica IX/metabolismo , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/metabolismo , Femenino , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Técnicas para Inmunoenzimas , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Invasividad Neoplásica , Estadificación de Neoplasias , Neovascularización Patológica , Estudios Retrospectivos
7.
Mol Pharm ; 13(2): 534-44, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26713599

RESUMEN

Fluorescence molecular imaging can be employed for the development of novel cancer targeting agents. Herein, we investigated the pharmacokinetics (PK) and cellular uptake of Dmt-Tic-Cy5, a delta-opioid receptor (δOR) antagonist-fluorescent dye conjugate, as a tumor-targeting molecular imaging agent. δOR expression is observed normally in the CNS, and pathologically in some tumors, including lung liver and breast cancers. In vitro, in vivo, and ex vivo experiments were conducted to image and quantify the fluorescence signal associated with Dmt-Tic-Cy5 over time using in vitro and intravital fluorescence microscopy and small animal fluorescence imaging of tumor-bearing mice. We observed specific retention of Dmt-Tic-Cy5 in tumors with maximum uptake in δOR-expressing positive tumors at 3 h and observable persistence for >96 h; clearance from δOR nonexpressing negative tumors by 6 h; and systemic clearance from normal organs by 24 h. Live-cell and intravital fluorescence microscopy demonstrated that Dmt-Tic-Cy5 had sustained cell-surface binding lasting at least 24 h with gradual internalization over the initial 6 h following administration. Dmt-Tic-Cy5 is a δOR-targeted agent that exhibits long-lasting and specific signal in δOR-expressing tumors, is rapidly cleared from systemic circulation, and is not retained in non-δOR-expressing tissues. Hence, Dmt-Tic-Cy5 has potential as a fluorescent tumor imaging agent.


Asunto(s)
Carbocianinas/farmacocinética , Neoplasias del Colon/tratamiento farmacológico , Dipéptidos/farmacocinética , Colorantes Fluorescentes/química , Receptores Opioides delta/química , Tetrahidroisoquinolinas/farmacocinética , Animales , Apoptosis , Carbocianinas/administración & dosificación , Proliferación Celular , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Dipéptidos/administración & dosificación , Femenino , Humanos , Técnicas para Inmunoenzimas , Cinética , Ratones , Ratones Desnudos , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/farmacocinética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectroscopía Infrarroja Corta , Tetrahidroisoquinolinas/administración & dosificación , Distribución Tisular , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Mol Imaging Biol ; 18(2): 219-31, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26276155

RESUMEN

PURPOSE: Hypoxia is commonly observed in regions of primary tumors and metastases, and is associated with resistance to treatment, more aggressive tumor phenotypes and poor prognosis. Reliable and validated imaging biomarkers of hypoxia are needed for pre-clinical studies and clinical use. Expression of cell-surface carbonic anhydrases IX and XII (CAIX and CAXII) in tumor cells has been associated with tumor hypoxia. CAIX and CAXII specific antibodies conjugated to fluorescent dye were evaluated for the non-invasive detection of hypoxia in vivo. PROCEDURES: Human breast cancer cell lines (MCF10A, DCIS, MCF7, ZR-75.1 and MDA-mb231) were characterized for CAIX and CAXII expression by real-time RT-PCR and immunocytochemistry (ICC) under normoxic and hypoxic conditions. Immunohistochemical (IHC) staining of CAIX, CAXII and the commercially available exogenous hypoxia marker, pimonidazole, was performed using sections of ZR-75.1 and MDA-mb-231 orthotopic breast cancer xenograft tumors from nude mice. In vivo fluorescence imaging of ZR-75.1 tumors in animals housed at varied levels of oxygen was used to quantify the relative uptake of the CAIX and CAXII agents and a commercially available sulfonamide-based agent. Corresponding tumor sections were IHC stained for CAIX, CAXII and pimonidazole. RESULTS: CAIX mRNA expression was significantly higher (p < 0.05) in hypoxia for all cell lines, which was in agreement with protein expression by ICC. CAXII expression was mixed, with a modest hypoxia-related increase in two cell lines (p < 0.05) and no change in others. Quantified IHC staining of ZR-75.1 and MDA-mb-231 tumor sections showed that CAIX and CAXII expression was elevated in regions with pimonidazole staining, but CAXII levels were lower than CAIX. Tumor uptake of the CAIX targeted agent, and IHC staining of CAIX and pimonidazole in corresponding tumor sections were correlated, and co-registered, and shown to be significantly elevated by level of oxygenation (p < 0.001): hypoxia > normoxia > hyperoxia. However, the CAXII and sulfonamide agents were not significantly correlated with hypoxia. CONCLUSION: These studies suggest that the fluorescently labeled CAIX-specific agent is a more robust indicator of hypoxia in vivo compared to the CAXII-specific agent or the agent specific to the CA active site.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/enzimología , Anhidrasa Carbónica IX/metabolismo , Anhidrasas Carbónicas/metabolismo , Imagen Molecular/métodos , Oxígeno/farmacología , Hipoxia Tumoral/efectos de los fármacos , Hipoxia Tumoral/genética , Análisis de Varianza , Animales , Antígenos de Neoplasias/genética , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Anhidrasa Carbónica IX/genética , Anhidrasas Carbónicas/genética , Línea Celular Tumoral , Femenino , Fluorescencia , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones Desnudos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tomografía , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Nat Commun ; 6: 8752, 2015 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-26658462

RESUMEN

Early cancers are avascular and hence, profoundly acidic. Pre-malignant cells must adapt to acidosis to thrive in this hostile microenvironment. Here, we investigate MCF-7 cells that are adapted to grow in acidic conditions using SILAC proteomics and we reveal a significant upregulation of lysosomal proteins. Prominent among these is LAMP2 that functions to protect lysosomal membranes from acid proteolysis. LAMP2 upregulation by acidosis is confirmed both in vitro and in vivo. Furthermore, we show that the depletion of LAMP2 is sufficient to increase acidosis-mediated toxicity. In breast cancer patient samples, there is a high correlation of LAMP2 mRNA and protein expression with progression. We also observe that LAMP2 is located at the plasma membrane in clinical samples and this redistribution is acid-induced in vitro. Our findings suggest a potential adaptive mechanism, wherein cells chronically exposed to an acidic environment translocate lysosomal proteins to their surface, thus protecting the plasmalemma from acid-induced hydrolysis.


Asunto(s)
Membrana Celular/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Neoplasias/metabolismo , Animales , Biomarcadores de Tumor , Línea Celular Tumoral , Femenino , Humanos , Concentración de Iones de Hidrógeno , Proteína 2 de la Membrana Asociada a los Lisosomas/genética , Ratones , Ratones Desnudos , Neoplasias Experimentales/metabolismo , Análisis por Matrices de Proteínas , Proteómica
10.
Adv Anat Pathol ; 22(4): 267-72, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26050264

RESUMEN

A major goal of modern medicine is increasing patient specificity so that the right treatment is administered to the right patient at the right time with the right dose. While current cancer studies have largely focused on identification of genetic or epigenetic properties of tumor cells, emerging evidence has clearly demonstrated substantial genetic heterogeneity between tumors in the same patient and within subclones of a single tumor. Thus, molecular analysis from populations of cells (either a whole tumor or small biopsy of that tumor) is, at best, an incomplete representation of the underlying biology. These observations indicate a significant need to define intratumoral evolutionary dynamics that yield the observed spatial variations in cellular properties. It is generally accepted that genetic heterogeneity among cancer cells is a manifestation of intratumoral evolution, and this is typically viewed as a consequence of random mutations generated by genomic instability within the cancer cells. We suggest that this represents an incomplete view of Darwinian dynamics, which typically are governed by phenotypic variations in response to spatial and temporal heterogeneity in environmental selection forces. We propose that pathologic feature analysis can provide precise information regarding regional variations in environmental selection forces and phenotypic adaptations. These observations can be integrated using quantitative, spatially explicit methods developed in landscape ecology to interrogate heterogenous biological processes in tumors within individual patients. The ability to investigate tumor heterogeneity has been shown to inform physicians regarding critical aspects of cancer progression including invasion, metastasis, drug resistance, and disease relapse.


Asunto(s)
Neoplasias/patología , Medicina de Precisión/métodos , Humanos , Patología
11.
J Leukoc Biol ; 97(4): 807-19, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25663681

RESUMEN

Th17 cells contribute to severe GVHD in murine bone marrow transplantation. Targeted deletion of the RORγt transcription factor or blockade of the JAK2-STAT3 axis suppresses IL-17 production and alloreactivity by Th17 cells. Here, we show that pSTAT3 Y705 is increased significantly in CD4(+) T cells among human recipients of allogeneic HCT before the onset of Grade II-IV acute GVHD. Examination of target-organ tissues at the time of GVHD diagnosis indicates that the amount of RORγt + Th17 cells is significantly higher in severe GVHD. Greater accumulation of tissue-resident Th17 cells also correlates with the use of MTX- compared with Rapa-based GVHD prophylaxis, as well as a poor therapeutic response to glucocorticoids. RORγt is optimally suppressed by concurrent neutralization of TORC1 with Rapa and inhibition of STAT3 activation with S3I-201, supporting that mTOR- and STAT3-dependent pathways converge upon RORγt gene expression. Rapa-resistant T cell proliferation can be totally inhibited by STAT3 blockade during initial allosensitization. We conclude that STAT3 signaling and resultant Th17 tissue accumulation are closely associated with acute GVHD onset, severity, and treatment outcome. Future studies are needed to validate the association of STAT3 activity in acute GVHD. Novel GVHD prevention strategies that incorporate dual STAT3 and mTOR inhibition merit investigation.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Enfermedad Injerto contra Huésped/inmunología , Procesamiento Proteico-Postraduccional , Factor de Transcripción STAT3/metabolismo , Células Th17/inmunología , Enfermedad Aguda , Adulto , Anciano , Aloinjertos , Ácidos Aminosalicílicos/farmacología , Bencenosulfonatos/farmacología , Trasplante de Médula Ósea , Células Dendríticas/inmunología , Femenino , Glucocorticoides/uso terapéutico , Enfermedad Injerto contra Huésped/sangre , Enfermedad Injerto contra Huésped/terapia , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/terapia , Humanos , Inmunosupresores/uso terapéutico , Prueba de Cultivo Mixto de Linfocitos , Masculino , Metotrexato/uso terapéutico , Persona de Mediana Edad , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/biosíntesis , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/sangre , Trasplante de Células Madre de Sangre Periférica , Fosforilación , Estudios Prospectivos , Receptores de Interleucina-6/sangre , Factor de Transcripción STAT3/antagonistas & inhibidores , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/fisiología
12.
Anticancer Res ; 35(1): 25-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25550531

RESUMEN

BACKGROUND/AIM: Melanoma cells express high levels of HLA class II, cell surface antigen-presenting proteins, which is an anomalous phenotype among solid tumors. There has never been a satisfying explanation for how this HLA class II-positive phenotype is related to tumor development. Lugini and colleagues demonstrated that melanoma cells have the capacity to engulf T-cells. We considered the possibility that this capacity could be dependent on HLA class II expression. MATERIALS AND METHODS: We co-cultured melanoma and CD4-positive, labeled, Jurkat-C T-cells. The melanoma cells were transformed with an expression vector for CIITA, the obligate HLA class II gene transactivator. We then assayed for the transfer of label to the melanoma cells. RESULTS: CIITA expression facilitated engulfment of the T-cell material but not material from B-cells. CONCLUSION: The results suggest a possible mechanism for HLA class II-positive melanoma cells in blunting an anti-tumor response and suggest a possible target for melanoma therapy.


Asunto(s)
Melanoma/inmunología , Proteínas Nucleares/metabolismo , Transactivadores/metabolismo , Técnicas de Cocultivo , Humanos , Células Jurkat , Melanoma/patología , Fagocitosis
13.
Cancer Metab ; 3(1): 2, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25635223

RESUMEN

BACKGROUND: Hypoxic niches in solid tumors harbor therapy-resistant cells. Hypoxia-activated prodrugs (HAPs) have been designed to overcome this resistance and, to date, have begun to show clinical efficacy. However, clinical HAPs activity could be improved. In this study, we sought to identify non-pharmacological methods to acutely exacerbate tumor hypoxia to increase TH-302 activity in pancreatic ductal adenocarcinoma (PDAC) tumor models. RESULTS: Three human PDAC cell lines with varying sensitivity to TH-302 (Hs766t > MiaPaCa-2 > SU.86.86) were used to establish PDAC xenograft models. PDAC cells were metabolically profiled in vitro and in vivo using the Seahorse XF system and hyperpolarized (13)C pyruvate MRI, respectively, in addition to quantitative immunohistochemistry. The effect of exogenous pyruvate on tumor oxygenation was determined using electroparamagnetic resonance (EPR) oxygen imaging. Hs766t and MiaPaCa-2 cells exhibited a glycolytic phenotype in comparison to TH-302 resistant line SU.86.86. Supporting this observation is a higher lactate/pyruvate ratio in Hs766t and MiaPaCa xenografts as observed during hyperpolarized pyruvate MRI studies in vivo. Coincidentally, response to exogenous pyruvate both in vitro (Seahorse oxygen consumption) and in vivo (EPR oxygen imaging) was greatest in Hs766t and MiaPaCa models, possibly due to a higher mitochondrial reserve capacity. Changes in oxygen consumption and in vivo hypoxic status to pyruvate were limited in the SU.86.86 model. Combination therapy of pyruvate plus TH-302 in vivo significantly decreased tumor growth and increased survival in the MiaPaCa model and improved survival in Hs766t tumors. CONCLUSIONS: Using metabolic profiling, functional imaging, and computational modeling, we show improved TH-302 activity by transiently increasing tumor hypoxia metabolically with exogenous pyruvate. Additionally, this work identified a set of biomarkers that may be used clinically to predict which tumors will be most responsive to pyruvate + TH-302 combination therapy. The results of this study support the concept that acute increases in tumor hypoxia can be beneficial for improving the clinical efficacy of HAPs and can positively impact the future treatment of PDAC and other cancers.

14.
PLoS One ; 10(1): e0116869, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25607660

RESUMEN

BACKGROUND: Intraductal papillary mucinous neoplasms (IPMNs) are pancreatic ductal adenocarcinoma (PDAC) precursors. Differentiating between high-risk IPMNs that warrant surgical resection and low-risk IPMNs that can be monitored is a significant clinical problem, and we sought to discover a panel of mi(cro)RNAs that accurately classify IPMN risk status. METHODOLOGY/PRINCIPAL FINDINGS: In a discovery phase, genome-wide miRNA expression profiling was performed on 28 surgically-resected, pathologically-confirmed IPMNs (19 high-risk, 9 low-risk) using Taqman MicroRNA Arrays. A validation phase was performed in 21 independent IPMNs (13 high-risk, 8 low-risk). We also explored associations between miRNA expression level and various clinical and pathological factors and examined genes and pathways regulated by the identified miRNAs by integrating data from bioinformatic analyses and microarray analysis of miRNA gene targets. Six miRNAs (miR-100, miR-99b, miR-99a, miR-342-3p, miR-126, miR-130a) were down-regulated in high-risk versus low-risk IPMNs and distinguished between groups (P<10-3, area underneath the curve (AUC) = 87%). The same trend was observed in the validation phase (AUC = 74%). Low miR-99b expression was associated with main pancreatic duct involvement (P = 0.021), and serum albumin levels were positively correlated with miR-99a (r = 0.52, P = 0.004) and miR-100 expression (r = 0.49, P = 0.008). Literature, validated miRNA:target gene interactions, and pathway enrichment analysis supported the candidate miRNAs as tumor suppressors and regulators of PDAC development. Microarray analysis revealed that oncogenic targets of miR-130a (ATG2B, MEOX2), miR-342-3p (DNMT1), and miR-126 (IRS-1) were up-regulated in high- versus low-risk IPMNs (P<0.10). CONCLUSIONS: This pilot study highlights miRNAs that may aid in preoperative risk stratification of IPMNs and provides novel insights into miRNA-mediated progression to pancreatic malignancy. The miRNAs identified here and in other recent investigations warrant evaluation in biofluids in a well-powered prospective cohort of individuals newly-diagnosed with IPMNs and other pancreatic cysts and those at increased genetic risk for these lesions.


Asunto(s)
Adenocarcinoma Mucinoso/patología , Adenocarcinoma Papilar/patología , Carcinoma Ductal Pancreático/patología , MicroARNs/genética , Neoplasias Pancreáticas/patología , Adenocarcinoma Mucinoso/genética , Adenocarcinoma Papilar/genética , Anciano , Anciano de 80 o más Años , Carcinoma Ductal Pancreático/genética , Diagnóstico Diferencial , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Neoplasias Pancreáticas/genética , Proyectos Piloto , Albúmina Sérica/metabolismo
15.
BMC Cancer ; 14: 279, 2014 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-24755315

RESUMEN

BACKGROUND: Breast carcinoma can be classified as either Estrogen Receptor (ER) positive or negative by immunohistochemical phenotyping, although ER expression may vary from 1 to 100% of malignant cells within an ER + tumor. This is similar to genetic variability observed in other tumor types and is generally viewed as a consequence of intratumoral evolution driven by random genetic mutations. Here we view cellular evolution within tumors as a classical Darwinian system in which variations in molecular properties represent predictable adaptations to spatially heterogeneous environmental selection forces. We hypothesize that ER expression is a successful adaptive strategy only if estrogen is present in the microenvironment. Since the dominant source of estrogen is blood flow, we hypothesized that, in general, intratumoral regions with higher blood flow would contain larger numbers of ER + cells when compared to areas of low blood flow and in turn necrosis. METHODS: This study used digital pathology whole slide image acquisition and advanced image analysis algorithms. We examined the spatial distribution of ER + and ER- cells, vascular density, vessel area, and tissue necrosis within histological sections of 24 breast cancer specimens. These data were correlated with the patients ER status and molecular pathology report findings. RESULTS: ANOVA analyses revealed a strong correlation between vascular area and ER expression and between high fractional necrosis and absent ER expression (R(2) = 39%; p < 0.003 and R(2) = 46%; p < 0.001), respectively). ER expression did not correlate with tumor grade or size. CONCLUSION: We conclude that ER expression can be understood as a Darwinian process and linked to variations in estrogen delivery by temporal and spatial heterogeneity in blood flow. This correlation suggests strategies to promote intratumoral blood flow or a cyclic introduction of estrogen in the treatment schedule could be explored as a counter-intuitive approach to increase the efficacy of anti-estrogen drugs.


Asunto(s)
Neoplasias de la Mama/genética , Receptor alfa de Estrógeno/biosíntesis , Estrógenos/genética , Selección Genética/genética , Neoplasias de la Mama/patología , Receptor alfa de Estrógeno/genética , Estrógenos/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Necrosis/genética , Necrosis/patología , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Microambiente Tumoral/genética
16.
Subcell Biochem ; 75: 221-54, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24146382

RESUMEN

Carbonic anhydrase IX (CAIX) which is a zinc containing metalloprotein, efficiently catalyzes the reversible hydration of carbon dioxide. It is constitutively up-regulated in several cancer types and has an important role in tumor progression, acidification and metastasis. High expression of CAIX generally correlates with poor prognosis and is related to a decrease in the disease-free interval following successful therapy. Therefore, it is considered as a prognostic indicator in oncology.In this review, we describe CAIX regulation and its role in tumor hypoxia, acidification and metastasis. In addition, the molecular imaging of CAIX and its potential for use in cancer detection, diagnosis, staging, and for use in following therapy response is discussed. Both antibodies and small molecular weight compounds have been used for targeted imaging of CAIX expression. The use of CAIX expression as an attractive and promising candidate marker for systemic anticancer therapy is also discussed.


Asunto(s)
Antígenos de Neoplasias/genética , Anhidrasas Carbónicas/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Antígenos de Neoplasias/biosíntesis , Dióxido de Carbono/metabolismo , Anhidrasa Carbónica IX , Anhidrasas Carbónicas/biosíntesis , Hipoxia de la Célula/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/patología , Zinc/metabolismo
17.
PLoS One ; 8(12): e82875, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24358232

RESUMEN

Combinations of targeted drugs have been employed to treat sarcomas, however, response rates have not improved notably, therefore emphasizing the need for novel treatments. In addition, imaging approaches to assess therapeutic response is lacking, as currently measurable indices, such as volume and/or diameter, do not accurately correlate with changes in tumor biology. In this study, quantitative and profound analyses of magnetic resonance imaging (MRI) were developed to evaluate these as imaging biomarkers for MK1775 and Gem in an osteosarcoma xenotransplant model at early time-points following treatment. Notably, we showed that Gem and Gem+MK1775 groups had significantly inhibited tumor growth by day 4, which was presaged by elevations in mean ADC by 24 hours post treatment. Significant differences were also observed at later time points for the Gem+MK1775 combination and MK1775 therapy. ADC distribution and entropy (randomness of ADC values) were also elevated by 24 hours following therapy. Immunohistochemistry demonstrated that these treatment-related increases in ADC correlated with apoptosis and observed cell condensations (dense- and exploded bodies). These findings underline the role of ADC as a quantitative imaging biomarker for therapy-induced response and show promising clinical relevance in the sarcoma patient population.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Óseas/diagnóstico , Neoplasias Óseas/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Imagen de Difusión por Resonancia Magnética , Osteosarcoma/diagnóstico , Osteosarcoma/tratamiento farmacológico , Pirazoles/administración & dosificación , Pirimidinas/administración & dosificación , Animales , Neoplasias Óseas/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Desoxicitidina/administración & dosificación , Femenino , Humanos , Ratones , Ratones SCID , Persona de Mediana Edad , Terapia Molecular Dirigida , Osteosarcoma/patología , Pronóstico , Pirimidinonas , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
18.
J Med Chem ; 56(16): 6330-8, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23863078

RESUMEN

Recent emphasis has focused on the development of rationally designed polymer-based micelle carriers for drug delivery. The current work tests the hypothesis that target specificity can be enhanced by micelles with cancer-specific ligands. In particular, we describe the synthesis and characterization of a new gadolinium texaphyrin (Gd-Tx) complex encapsulated in an IVECT micellar system, stabilized through Fe(III) cross-linking and targeted with multiple copies of a specific ligand for the melanocortin 1 receptor (MC1R), which has been evaluated as a cell-surface marker for melanoma. On the basis of comparative MRI experiments, we have been able to demonstrate that these Gd-Tx micelles are able to target MC1R-expressing xenograft tumors in vitro and in vivo more effectively than various control systems, including untargeted or un-cross-linked Gd-Tx micelles. Taken in concert, the findings reported herein support the conclusion that appropriately designed micelles are able to deliver contrast agent payloads to tumors expressing the MC1R.


Asunto(s)
Imagen por Resonancia Magnética/métodos , Micelas , Polímeros/química , Receptor de Melanocortina Tipo 1/química , Animales , Línea Celular Tumoral , Humanos , Ratones , Ratones SCID
19.
Mol Pharm ; 10(8): 3175-85, 2013 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-23763620

RESUMEN

The melanocortin 1 receptor (MC1R) is overexpressed in most melanoma metastases, making it a promising target for imaging of melanomas. In this study, the expression of MC1R in a large fraction of patients with melanoma was confirmed using mRNA and tissue microarray. Here, we have characterized the in vivo tumor and tissue distribution and pharmacokinetics (PK) of uptake and clearance of a MC1R specific peptidomimetic ligand conjugated to a near-infrared fluorescent dye. We propose an interdisciplinary framework to bridge the different time and space scales of ligand-tumor-host interactions: intravital fluorescence microscopy to quantify probe internalization at the cellular level, a xenograft tumor model for whole body pharmacokinetics, and a computational pharmacokinetic model for integration and interpretation of experimental data. Administration of the probe into mice bearing tumors with high and low MC1R expression demonstrated normalized image intensities that correlated with expression levels (p < 0.05). The biodistribution study showed high kidney uptake as early as 30 min postinjection. The PK computational model predicted the presence of receptors in the kidneys with a lower affinity, but at higher numbers than in the tumors. As the mouse kidney is known to express the MC5R, this hypothesis was confirmed by both coinjection of a ligand with higher MC5R affinity compared to MC1R and by injection of lower probe concentrations (e.g., 1 nmol/kg), both leading to decreased kidney accumulation of the MC1R ligand. In addition, through this interdisciplinary approach we could predict the rates of ligand accumulation and clearance into and from organs and tumors, and the amount of injected ligand required to have maximum specific retention in tumors. These predictions have potential to aid in the translation of a targeted agent from lab to the clinic. In conclusion, the characterized MC1R-specific probe has excellent potential for in vivo detection of melanoma metastases. The process of cell-surface marker validation, targeted imaging probe development, and in vitro, in vivo, and in silico characterization described in this study can be generally applied to preclinical development of targeted agents.


Asunto(s)
Melanoma/metabolismo , Sondas Moleculares/metabolismo , Sondas Moleculares/farmacocinética , Receptor de Melanocortina Tipo 1/metabolismo , Animales , Línea Celular Tumoral , Femenino , Humanos , Inmunohistoquímica , Ratones , Modelos Teóricos , Receptor de Melanocortina Tipo 1/genética , Análisis de Matrices Tisulares , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Front Oncol ; 3: 111, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23717812

RESUMEN

Despite the great progress that has been made in understanding cancer biology and the potential molecular targets for its treatment, the majority of drugs fail in the clinical trials. This may be attributed (at least in part) to the complexity of interstitial drug transport in the patient's body, which is hard to test experimentally. Similarly, recent advances in molecular imaging have led to the development of targeted biomarkers that can predict pharmacological responses to therapeutic interventions. However, both the drug and biomarker molecules need to access the tumor tissue and be taken up into individual cells in concentrations sufficient to exert the desired effect. To investigate the process of drug penetration at the mesoscopic level we developed a computational model of interstitial transport that incorporates the biophysical properties of the tumor tissue, including its architecture and interstitial fluid flow, as well as the properties of the agents. This model is based on the method of regularized Stokeslets to describe the fluid flow coupled with discrete diffusion-advection-reaction equations to model the dynamics of the drugs. Our results show that the tissue cellular porosity and density influence the depth of penetration in a non-linear way, with sparsely packed tissues being traveled through more slowly than the denser tissues. We demonstrate that irregularities in the cell spatial configurations result in the formation of interstitial corridors that are followed by agents leading to the emergence of tissue zones with less exposure to the drugs. We describe how the model can be integrated with in vivo experiments to test the extravasation and penetration of the targeted biomarkers through the tumor tissue. A better understanding of tissue- or compound-specific factors that limit the penetration through the tumors is important for non-invasive diagnoses, chemotherapy, the monitoring of treatment responses, and the detection of tumor recurrence.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA