Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(34): e2220269120, 2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37579172

RESUMEN

The vascular endothelium from individual organs is functionally specialized, and it displays a unique set of accessible molecular targets. These serve as endothelial cell receptors to affinity ligands. To date, all identified vascular receptors have been proteins. Here, we show that an endothelial lung-homing peptide (CGSPGWVRC) interacts with C16-ceramide, a bioactive sphingolipid that mediates several biological functions. Upon binding to cell surfaces, CGSPGWVRC triggers ceramide-rich platform formation, activates acid sphingomyelinase and ceramide production, without the associated downstream apoptotic signaling. We also show that the lung selectivity of CGSPGWVRC homing peptide is dependent on ceramide production in vivo. Finally, we demonstrate two potential applications for this lipid vascular targeting system: i) as a bioinorganic hydrogel for pulmonary imaging and ii) as a ligand-directed lung immunization tool against COVID-19. Thus, C16-ceramide is a unique example of a lipid-based receptor system in the lung vascular endothelium targeted in vivo by circulating ligands such as CGSPGWVRC.


Asunto(s)
COVID-19 , Humanos , Ligandos , COVID-19/metabolismo , Ceramidas/metabolismo , Pulmón/metabolismo , Endotelio Vascular/metabolismo , Receptores de Superficie Celular/metabolismo , Proteínas Portadoras/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo
2.
Mol Biol Evol ; 39(5)2022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35511693

RESUMEN

Evaluation of immunogenic epitopes for universal vaccine development in the face of ongoing SARS-CoV-2 evolution remains a challenge. Herein, we investigate the genetic and structural conservation of an immunogenically relevant epitope (C662-C671) of spike (S) protein across SARS-CoV-2 variants to determine its potential utility as a broad-spectrum vaccine candidate against coronavirus diseases. Comparative sequence analysis, structural assessment, and molecular dynamics simulations of C662-C671 epitope were performed. Mathematical tools were employed to determine its mutational cost. We found that the amino acid sequence of C662-C671 epitope is entirely conserved across the observed major variants of SARS-CoV-2 in addition to SARS-CoV. Its conformation and accessibility are predicted to be conserved, even in the highly mutated Omicron variant. Costly mutational rate in the context of energy expenditure in genome replication and translation can explain this strict conservation. These observations may herald an approach to developing vaccine candidates for universal protection against emergent variants of coronavirus.


Asunto(s)
COVID-19 , Vacunas , Epítopos de Linfocito T/química , Epítopos de Linfocito T/genética , Humanos , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética
3.
Bioessays ; 43(7): e2000339, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33751590

RESUMEN

Here we review and discuss the link between regeneration capacity and tumor suppression comparing mammals (embryos versus adults) with highly regenerative vertebrates. Similar to mammal embryo morphogenesis, in amphibians (essentially newts and salamanders) the reparative process relies on a precise molecular and cellular machinery capable of sensing abnormal signals and actively reprograming or eliminating them. As the embryo's evil twin, tumor also retains common functional attributes. The immune system plays a pivotal role in maintaining a physiological balance to provide surveillance against tumor initiation or to support its initiation and progression. We speculate that susceptibility to cancer development in adult mammals may be determined by the loss of an advanced regenerative capability during evolution and believe that gaining mechanistic insights into how regenerative capacity linked to tumor suppression is postnatally lost in mammals might illuminate an as yet unrecognized route to cancer treatment.


Asunto(s)
Anfibios , Neoplasias , Animales , Biología , Embrión de Mamíferos , Humanos , Mamíferos , Neoplasias/genética
4.
Langmuir ; 38(45): 13983-13994, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36318182

RESUMEN

Delivery of small molecules and anticancer agents to malignant cells or specific regions within a tumor is limited by penetration depth and poor spatial drug distribution, hindering anticancer efficacy. Herein, we demonstrate control over gold nanoparticle (GNP) penetration and spatial distribution across solid tumors by administering GNPs with different surface chemistries at a constant injection rate via syringe pump. A key finding in this study is the discovery of different zone-specific accumulation patterns of intratumorally injected nanoparticles dependent on surface functionalization. Computed tomography (CT) imaging performed in vivo of C57BL/6 mice harboring Lewis lung carcinoma (LLC) tumors on their flank and gross visualization of excised tumors consistently revealed that intratumorally administered citrate-GNPs accumulate in particle clusters in central areas of the tumor, while GNPs functionalized with thiolated phosphothioethanol (PTE-GNPs) and thiolated polyethylene glycol (PEG-GNPs) regularly accumulate in the tumor periphery. Further, PEG functionalization resulted in larger tumoral surface coverage than PTE, reaching beyond the outer zone of the tumor mass and into the surrounding stroma. To understand the dissimilarities in spatiotemporal evolution across the different GNP surface chemistries, we modeled their intratumoral transport with reaction-diffusion equations. Our results suggest that GNP surface passivation affects nanoparticle reactivity with the tumor microenvironment, leading to differential transport behavior across tumor zones. The present study provides a mechanistic understanding of the factors affecting spatiotemporal distribution of nanoparticles in the tumor. Our proof of concept of zonal delivery within the tumor may prove useful for directing anticancer therapies to regions of biomarker overexpression.


Asunto(s)
Nanopartículas del Metal , Nanopartículas , Animales , Ratones , Oro , Ratones Endogámicos C57BL , Polietilenglicoles , Ácido Cítrico
5.
Pharm Res ; 39(3): 511-528, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35294699

RESUMEN

PURPOSE: Downregulation of miRNA-22 in triple-negative breast cancer (TNBC) is associated with upregulation of eukaryotic elongation 2 factor kinase (eEF2K) protein, which regulates tumor growth, chemoresistance, and tumor immunosurveillance. Moreover, exogenous administration of miRNA-22, loaded in nanoparticles to prevent degradation and improve tumor delivery (termed miRNA-22 nanotherapy), to suppress eEF2K production has shown potential as an investigational therapeutic agent in vivo. METHODS: To evaluate the translational potential of miRNA-22 nanotherapy, we developed a multiscale mechanistic model, calibrated to published in vivo data and extrapolated to the human scale, to describe and quantify the pharmacokinetics and pharmacodynamics of miRNA-22 in virtual patient populations. RESULTS: Our analysis revealed the dose-response relationship, suggested optimal treatment frequency for miRNA-22 nanotherapy, and highlighted key determinants of therapy response, from which combination with immune checkpoint inhibitors was identified as a candidate strategy for improving treatment outcomes. More importantly, drug synergy was identified between miRNA-22 and standard-of-care drugs against TNBC, providing a basis for rational therapeutic combinations for improved response CONCLUSIONS: The present study highlights the translational potential of miRNA-22 nanotherapy for TNBC in combination with standard-of-care drugs.


Asunto(s)
MicroARNs , Nanopartículas , Neoplasias de la Mama Triple Negativas , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , MicroARNs/administración & dosificación , MicroARNs/genética , Nanopartículas/administración & dosificación , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética
6.
J Am Chem Soc ; 143(20): 7655-7670, 2021 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-33988982

RESUMEN

Aptamers, synthetic single-strand oligonucleotides that are similar in function to antibodies, are promising as therapeutics because of their minimal side effects. However, the stability and bioavailability of the aptamers pose a challenge. We developed aptamers converted from RNA aptamer to modified DNA aptamers that target phospho-AXL with improved stability and bioavailability. On the basis of the comparative analysis of a library of 17 converted modified DNA aptamers, we selected aptamer candidates, GLB-G25 and GLB-A04, that exhibited the highest bioavailability, stability, and robust antitumor effect in in vitro experiments. Backbone modifications such as thiophosphate or dithiophosphate and a covalent modification of the 5'-end of the aptamer with polyethylene glycol optimized the pharmacokinetic properties, improved the stability of the aptamers in vivo by reducing nuclease hydrolysis and renal clearance, and achieved high and sustained inhibition of AXL at a very low dose. Treatment with these modified aptamers in ovarian cancer orthotopic mouse models significantly reduced tumor growth and the number of metastases. This effective silencing of the phospho-AXL target thus demonstrated that aptamer specificity and bioavailability can be improved by the chemical modification of existing aptamers for phospho-AXL. These results lay the foundation for the translation of these aptamer candidates and companion biomarkers to the clinic.


Asunto(s)
Anticuerpos/inmunología , Aptámeros de Nucleótidos/inmunología , Neoplasias/inmunología , Anticuerpos/química , Aptámeros de Nucleótidos/química , Humanos , Neoplasias/terapia
7.
Gut ; 69(10): 1818-1831, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31988194

RESUMEN

OBJECTIVE: To investigate the function of a novel primate-specific long non-coding RNA (lncRNA), named FLANC, based on its genomic location (co-localised with a pyknon motif), and to characterise its potential as a biomarker and therapeutic target. DESIGN: FLANC expression was analysed in 349 tumours from four cohorts and correlated to clinical data. In a series of multiple in vitro and in vivo models and molecular analyses, we characterised the fundamental biological roles of this lncRNA. We further explored the therapeutic potential of targeting FLANC in a mouse model of colorectal cancer (CRC) metastases. RESULTS: FLANC, a primate-specific lncRNA feebly expressed in normal colon cells, was significantly upregulated in cancer cells compared with normal colon samples in two independent cohorts. High levels of FLANC were associated with poor survival in two additional independent CRC patient cohorts. Both in vitro and in vivo experiments demonstrated that the modulation of FLANC expression influenced cellular growth, apoptosis, migration, angiogenesis and metastases formation ability of CRC cells. In vivo pharmacological targeting of FLANC by administration of 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine nanoparticles loaded with a specific small interfering RNA, induced significant decrease in metastases, without evident tissue toxicity or pro-inflammatory effects. Mechanistically, FLANC upregulated and prolonged the half-life of phosphorylated STAT3, inducing the overexpression of VEGFA, a key regulator of angiogenesis. CONCLUSIONS: Based on our findings, we discovered, FLANC as a novel primate-specific lncRNA that is highly upregulated in CRC cells and regulates metastases formation. Targeting primate-specific transcripts such as FLANC may represent a novel and low toxic therapeutic strategy for the treatment of patients.


Asunto(s)
Carcinogénesis , Proliferación Celular , Neoplasias Colorrectales , Neovascularización Patológica , ARN Largo no Codificante , Factor de Transcripción STAT3/metabolismo , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/terapia , Descubrimiento de Drogas , Regulación Neoplásica de la Expresión Génica , Marcadores Genéticos , Terapia Genética , Humanos , Ratones , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Pruebas de Farmacogenómica , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
Small ; 15(46): e1903747, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31565854

RESUMEN

Rapid sequestration and prolonged retention of intravenously injected nanoparticles by the liver and spleen (reticuloendothelial system (RES)) presents a major barrier to effective delivery to the target site and hampers clinical translation of nanomedicine. Inspired by biological macromolecular drugs, synthesis of ultrasmall (diameter ≈12-15 nm) porous silica nanoparticles (UPSNs), capable of prolonged plasma half-life, attenuated RES sequestration, and accelerated hepatobiliary clearance, is reported. The study further investigates the effect of tumor vascularization on uptake and retention of UPSNs in two mouse models of triple negative breast cancer with distinctly different microenvironments. A semimechanistic mathematical model is developed to gain mechanistic insights into the interactions between the UPSNs and the biological entities of interest, specifically the RES. Despite similar systemic pharmacokinetic profiles, UPSNs demonstrate strikingly different tumor responses in the two models. Histopathology confirms the differences in vasculature and stromal status of the two models, and corresponding differences in the microscopic distribution of UPSNs within the tumors. The studies demonstrate the successful application of multidisciplinary and complementary approaches, based on laboratory experimentation and mathematical modeling, to concurrently design optimized nanomaterials, and investigate their complex biological interactions, in order to drive innovation and translation.


Asunto(s)
Nanopartículas/química , Neovascularización Patológica/patología , Tamaño de la Partícula , Dióxido de Silicio/química , Neoplasias de la Mama Triple Negativas/irrigación sanguínea , Animales , Línea Celular Tumoral , Radioisótopos de Cobre/farmacocinética , Femenino , Humanos , Ratones Endogámicos BALB C , Modelos Biológicos , Nanopartículas/ultraestructura , Porosidad , Distribución Tisular , Neoplasias de la Mama Triple Negativas/diagnóstico por imagen , Neoplasias de la Mama Triple Negativas/patología , Microambiente Tumoral
9.
Biomed Microdevices ; 21(2): 40, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30949850

RESUMEN

Cancer continues to be among the leading healthcare problems worldwide, and efforts continue not just to find better drugs, but also better drug delivery methods. The need for delivering cytotoxic agents selectively to cancerous cells, for improved safety and efficacy, has triggered the application of nanotechnology in medicine. This effort has provided drug delivery systems that can potentially revolutionize cancer treatment. Nanocarriers, due to their capacity for targeted drug delivery, can shift the balance of cytotoxicity from healthy to cancerous cells. The field of cancer nanomedicine has made significant progress, but challenges remain that impede its clinical translation. Several biophysical barriers to the transport of nanocarriers to the tumor exist, and a much deeper understanding of nano-bio interactions is necessary to change the status quo. Mathematical modeling has been instrumental in improving our understanding of the physicochemical and physiological underpinnings of nanomaterial behavior in biological systems. Here, we present a comprehensive review of literature on mathematical modeling works that have been and are being employed towards a better understanding of nano-bio interactions for improved tumor delivery efficacy.


Asunto(s)
Modelos Biológicos , Nanomedicina , Neoplasias , Animales , Transporte Biológico , Humanos , Nanopartículas/química , Neoplasias/metabolismo , Distribución Tisular
10.
Proc Natl Acad Sci U S A ; 113(7): 1877-82, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26839407

RESUMEN

A major challenge of targeted molecular imaging and drug delivery in cancer is establishing a functional combination of ligand-directed cargo with a triggered release system. Here we develop a hydrogel-based nanotechnology platform that integrates tumor targeting, photon-to-heat conversion, and triggered drug delivery within a single nanostructure to enable multimodal imaging and controlled release of therapeutic cargo. In proof-of-concept experiments, we show a broad range of ligand peptide-based applications with phage particles, heat-sensitive liposomes, or mesoporous silica nanoparticles that self-assemble into a hydrogel for tumor-targeted drug delivery. Because nanoparticles pack densely within the nanocarrier, their surface plasmon resonance shifts to near-infrared, thereby enabling a laser-mediated photothermal mechanism of cargo release. We demonstrate both noninvasive imaging and targeted drug delivery in preclinical mouse models of breast and prostate cancer. Finally, we applied mathematical modeling to predict and confirm tumor targeting and drug delivery. These results are meaningful steps toward the design and initial translation of an enabling nanotechnology platform with potential for broad clinical applications.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/tratamiento farmacológico , Modelos Animales de Enfermedad , Imagen Multimodal , Nanotecnología , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Sistemas de Liberación de Medicamentos , Femenino , Rayos Infrarrojos , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Resonancia por Plasmón de Superficie
11.
PLoS Comput Biol ; 12(4): e1004839, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27115287

RESUMEN

Mathematics is often used to model biological systems. In mammary gland development, mathematical modeling has been limited to acinar and branching morphogenesis and breast cancer, without reference to normal duct formation. We present a model of ductal elongation that exploits the geometrically-constrained shape of the terminal end bud (TEB), the growing tip of the duct, and incorporates morphometrics, region-specific proliferation and apoptosis rates. Iterative model refinement and behavior analysis, compared with biological data, indicated that the traditional metric of nipple to the ductal front distance, or percent fat pad filled to evaluate ductal elongation rate can be misleading, as it disregards branching events that can reduce its magnitude. Further, model driven investigations of the fates of specific TEB cell types confirmed migration of cap cells into the body cell layer, but showed their subsequent preferential elimination by apoptosis, thus minimizing their contribution to the luminal lineage and the mature duct.


Asunto(s)
Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/crecimiento & desarrollo , Modelos Biológicos , Animales , Apoptosis , Tipificación del Cuerpo , Ciclo Celular , Proliferación Celular , Tamaño de la Célula , Biología Computacional , Femenino , Cinética , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Modelos Anatómicos , Maduración Sexual
12.
PLoS Comput Biol ; 12(6): e1004969, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27286441

RESUMEN

AUTHOR SUMMARY: Cancer treatment efficacy can be significantly enhanced through the elution of drug from nano-carriers that can temporarily stay in the tumor vasculature. Here we present a relatively simple yet powerful mathematical model that accounts for both spatial and temporal heterogeneities of drug dosing to help explain, examine, and prove this concept. We find that the delivery of systemic chemotherapy through a certain form of nano-carriers would have enhanced tumor kill by a factor of 2 to 4 over the standard therapy that the patients actually received. We also find that targeting blood volume fraction (a parameter of the model) through vascular normalization can achieve more effective drug delivery and tumor kill. More importantly, this model only requires a limited number of parameters which can all be readily assessed from standard clinical diagnostic measurements (e.g., histopathology and CT). This addresses an important challenge in current translational research and justifies further development of the model towards clinical translation.


Asunto(s)
Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Modelos Biológicos , Neoplasias/tratamiento farmacológico , Animales , Biología Computacional , Simulación por Computador , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/uso terapéutico , Femenino , Ratones , Ratones Endogámicos BALB C , Nanopartículas/uso terapéutico , Análisis Espacio-Temporal
13.
Semin Cancer Biol ; 30: 70-8, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24793698

RESUMEN

There have been many techniques developed in recent years to in silico model a variety of cancer behaviors. Agent-based modeling is a specific discrete-based hybrid modeling approach that allows simulating the role of diversity in cell populations as well as within each individual cell; it has therefore become a powerful modeling method widely used by computational cancer researchers. Many aspects of tumor morphology including phenotype-changing mutations, the adaptation to microenvironment, the process of angiogenesis, the influence of extracellular matrix, reactions to chemotherapy or surgical intervention, the effects of oxygen and nutrient availability, and metastasis and invasion of healthy tissues have been incorporated and investigated in agent-based models. In this review, we introduce some of the most recent agent-based models that have provided insight into the understanding of cancer growth and invasion, spanning multiple biological scales in time and space, and we further describe several experimentally testable hypotheses generated by those models. We also discuss some of the current challenges of multiscale agent-based cancer models.


Asunto(s)
Modelos Biológicos , Modelos Teóricos , Neoplasias/patología , Investigación Biomédica Traslacional , Simulación por Computador , Humanos
14.
J Theor Biol ; 407: 259-270, 2016 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-27475843

RESUMEN

Mammary gland ductal elongation is spearheaded by terminal end buds (TEBs), where populations of highly proliferative cells are maintained throughout post-pubertal organogenesis in virgin mice until the mammary fat pad is filled by a mature ductal tree. We have developed a hybrid multiscale agent-based model to study how cellular differentiation pathways, cellular proliferation capacity, and endocrine and paracrine signaling play a role during development of the mammary gland. A simplified cellular phenotypic hierarchy that includes stem, progenitor, and fully differentiated cells within the TEB was implemented. Model analysis finds that mammary gland development was highly sensitive to proliferation events within the TEB, with progenitors likely undergoing 2-3 proliferation cycles before transitioning to a non-proliferative phenotype, and this result is in agreement with our previous experimental work. Endocrine and paracrine signaling were found to provide reliable ductal elongation rate regulation, while variations in the probability a new daughter cell will be of a proliferative phenotype were seen to have minimal effects on ductal elongation rates. Moreover, the distribution of cellular phenotypes within the TEB was highly heterogeneous, demonstrating significant allowable plasticity in possible phenotypic distributions while maintaining biologically relevant growth behavior. Finally, simulation results indicate ductal elongation rates due to cellular proliferation within the TEB may have a greater sensitivity to upstream endocrine signaling than endothelial to stromal paracrine signaling within the TEB. This model provides a useful tool to gain quantitative insights into cellular population dynamics and the effects of endocrine and paracrine signaling within the pubertal terminal end bud.


Asunto(s)
Glándulas Mamarias Animales/crecimiento & desarrollo , Análisis de Sistemas , Anfirregulina/metabolismo , Animales , División Celular Asimétrica , Ciclo Celular , Diferenciación Celular , Proliferación Celular , Estrógenos/metabolismo , Femenino , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Ratones , Fenotipo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo
15.
Proc Natl Acad Sci U S A ; 110(35): 14266-71, 2013 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-23940372

RESUMEN

Physical properties of the microenvironment influence penetration of drugs into tumors. Here, we develop a mathematical model to predict the outcome of chemotherapy based on the physical laws of diffusion. The most important parameters in the model are the volume fraction occupied by tumor blood vessels and their average diameter. Drug delivery to cells, and kill thereof, are mediated by these microenvironmental properties and affected by the diffusion penetration distance after extravasation. To calculate parameter values we fit the model to histopathology measurements of the fraction of tumor killed after chemotherapy in human patients with colorectal cancer metastatic to liver (coefficient of determination R(2) = 0.94). To validate the model in a different tumor type, we input patient-specific model parameter values from glioblastoma; the model successfully predicts extent of tumor kill after chemotherapy (R(2) = 0.7-0.91). Toward prospective clinical translation, we calculate blood volume fraction parameter values from in vivo contrast-enhanced computed tomography imaging from a separate cohort of patients with colorectal cancer metastatic to liver, and demonstrate accurate model predictions of individual patient responses (average relative error = 15%). Here, patient-specific data from either in vivo imaging or histopathology drives output of the model's formulas. Values obtained from standard clinical diagnostic measurements for each individual are entered into the model, producing accurate predictions of tumor kill after chemotherapy. Clinical translation will enable the rational design of individualized treatment strategies such as amount, frequency, and delivery platform of drug and the need for ancillary non-drug-based treatment.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Colorrectales/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Microambiente Tumoral , Neoplasias Encefálicas/patología , Neoplasias Colorrectales/patología , Glioblastoma/patología , Humanos , Perfusión , Estudios Prospectivos
16.
J Pharmacokinet Pharmacodyn ; 42(2): 179-89, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25588379

RESUMEN

Mathematical modeling has become a valuable tool that strives to complement conventional biomedical research modalities in order to predict experimental outcome, generate new medical hypotheses, and optimize clinical therapies. Two specific approaches, pharmacokinetic-pharmacodynamic (PK-PD) modeling, and agent-based modeling (ABM), have been widely applied in cancer research. While they have made important contributions on their own (e.g., PK-PD in examining chemotherapy drug efficacy and resistance, and ABM in describing and predicting tumor growth and metastasis), only a few groups have started to combine both approaches together in an effort to gain more insights into the details of drug dynamics and the resulting impact on tumor growth. In this review, we focus our discussion on some of the most recent modeling studies building on a combined PK-PD and ABM approach that have generated experimentally testable hypotheses. Some future directions are also discussed.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Neoplasias/tratamiento farmacológico , Humanos , Modelos Teóricos
17.
Phys Biol ; 11(6): 065002, 2014 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-25427073

RESUMEN

There is substantial heterogeneity in the clinical behavior of pancreatic cancer and in its response to therapy. Some of this variation may be due to differences in delivery of cytotoxic therapies between patients and within individual tumors. Indeed, in 12 patients with resectable pancreatic cancer, we previously demonstrated wide inter-patient variability in the delivery of gemcitabine as well as in the mass transport properties of tumors as measured by computed tomography (CT) scans. However, the variability of drug delivery and transport properties within pancreatic tumors is currently unknown. Here, we analyzed regional measurements of gemcitabine DNA incorporation in the tumors of the same 12 patients to understand the degree of intra-tumoral heterogeneity of drug delivery. We also developed a volumetric segmentation approach to measure mass transport properties from the CT scans of these patients and tested inter-observer agreement with this new methodology. Our results demonstrate significant heterogeneity of gemcitabine delivery within individual pancreatic tumors and across the patient cohort, with gemcitabine DNA incorporation in the inner portion of the tumors ranging from 38 to 74% of the total. Similarly, the CT-derived mass transport properties of the tumors had a high degree of heterogeneity, ranging from minimal difference to almost 200% difference between inner and outer portions of the tumor. Our quantitative method to derive transport properties from CT scans demonstrated less than 5% difference in gemcitabine prediction at the average CT-derived transport value across observers. These data illustrate significant inter-patient and intra-tumoral heterogeneity in the delivery of gemcitabine, and highlight how this variability can be reproducibly accounted for using principles of mass transport. With further validation as a biophysical marker, transport properties of tumors may be useful in patient selection for therapy and prediction of therapeutic outcome.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacocinética , Desoxicitidina/análogos & derivados , Sistemas de Liberación de Medicamentos , Neoplasias Pancreáticas/metabolismo , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/metabolismo , Transporte Biológico , Aductos de ADN/metabolismo , ADN de Neoplasias/metabolismo , Desoxicitidina/administración & dosificación , Desoxicitidina/metabolismo , Desoxicitidina/farmacocinética , Humanos , Inyecciones Intravenosas , Pancreatectomía , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , Distribución Tisular , Tomógrafos Computarizados por Rayos X , Microambiente Tumoral , Gemcitabina
18.
J Theor Biol ; 355: 194-207, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-24751927

RESUMEN

Vascularized tumor growth is characterized by both abnormal interstitial fluid flow and the associated interstitial fluid pressure (IFP). Here, we study the effect that these conditions have on the transport of therapeutic agents during chemotherapy. We apply our recently developed vascular tumor growth model which couples a continuous growth component with a discrete angiogenesis model to show that hypertensive IFP is a physical barrier that may hinder vascular extravasation of agents through transvascular fluid flux convection, which drives the agents away from the tumor. This result is consistent with previous work using simpler models without blood flow or lymphatic drainage. We consider the vascular/interstitial/lymphatic fluid dynamics to show that tumors with larger lymphatic resistance increase the agent concentration more rapidly while also experiencing faster washout. In contrast, tumors with smaller lymphatic resistance accumulate less agents but are able to retain them for a longer time. The agent availability (area-under-the curve, or AUC) increases for less permeable agents as lymphatic resistance increases, and correspondingly decreases for more permeable agents. We also investigate the effect of vascular pathologies on agent transport. We show that elevated vascular hydraulic conductivity contributes to the highest AUC when the agent is less permeable, but to lower AUC when the agent is more permeable. We find that elevated interstitial hydraulic conductivity contributes to low AUC in general regardless of the transvascular agent transport capability. We also couple the agent transport with the tumor dynamics to simulate chemotherapy with the same vascularized tumor under different vascular pathologies. We show that tumors with an elevated interstitial hydraulic conductivity alone require the strongest dosage to shrink. We further show that tumors with elevated vascular hydraulic conductivity are more hypoxic during therapy and that the response slows down as the tumor shrinks due to the heterogeneity and low concentration of agents in the tumor interior compared with the cases where other pathological effects may combine to flatten the IFP and thus reduce the heterogeneity. We conclude that dual normalizations of the micronevironment - both the vasculature and the interstitium - are needed to maximize the effects of chemotherapy, while normalization of only one of these may be insufficient to overcome the physical resistance and may thus lead to sub-optimal outcomes.


Asunto(s)
Antineoplásicos , Presión Sanguínea , Linfangiogénesis , Modelos Biológicos , Modelos Teóricos , Neoplasias , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Transporte Biológico Activo , Humanos , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/psicología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/fisiopatología
19.
PLoS Comput Biol ; 9(3): e1003008, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23555235

RESUMEN

Non-Hodgkin's lymphoma is a disseminated, highly malignant cancer, with resistance to drug treatment based on molecular- and tissue-scale characteristics that are intricately linked. A critical element of molecular resistance has been traced to the loss of functionality in proteins such as the tumor suppressor p53. We investigate the tissue-scale physiologic effects of this loss by integrating in vivo and immunohistological data with computational modeling to study the spatiotemporal physical dynamics of lymphoma growth. We compare between drug-sensitive Eµ-myc Arf-/- and drug-resistant Eµ-myc p53-/- lymphoma cell tumors grown in live mice. Initial values for the model parameters are obtained in part by extracting values from the cellular-scale from whole-tumor histological staining of the tumor-infiltrated inguinal lymph node in vivo. We compare model-predicted tumor growth with that observed from intravital microscopy and macroscopic imaging in vivo, finding that the model is able to accurately predict lymphoma growth. A critical physical mechanism underlying drug-resistant phenotypes may be that the Eµ-myc p53-/- cells seem to pack more closely within the tumor than the Eµ-myc Arf-/- cells, thus possibly exacerbating diffusion gradients of oxygen, leading to cell quiescence and hence resistance to cell-cycle specific drugs. Tighter cell packing could also maintain steeper gradients of drug and lead to insufficient toxicity. The transport phenomena within the lymphoma may thus contribute in nontrivial, complex ways to the difference in drug sensitivity between Eµ-myc Arf-/- and Eµ-myc p53-/- tumors, beyond what might be solely expected from loss of functionality at the molecular scale. We conclude that computational modeling tightly integrated with experimental data gives insight into the dynamics of Non-Hodgkin's lymphoma and provides a platform to generate confirmable predictions of tumor growth.


Asunto(s)
Biología Computacional/métodos , Linfoma no Hodgkin/patología , Modelos Biológicos , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Calibración , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclofosfamida/farmacología , Progresión de la Enfermedad , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Inmunohistoquímica , Linfoma no Hodgkin/tratamiento farmacológico , Linfoma no Hodgkin/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Patológica , Proteína p53 Supresora de Tumor/genética
20.
Nanomedicine ; 10(7): 1487-96, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24262998

RESUMEN

The abnormal tumor vasculature presents a major challenge to the adequate delivery of chemotherapeutics, often limiting efficacy. We developed a nanoparticle-based technique to deliver localized mild hyperthermia (MHT) used to transiently alter tumor vascular transport properties and enhance transport of macromolecules into tumor interstitium. The strategy involved administering and localizing accumulation of stealth gold nanorods (GNRs, 103 µg of GNRs/g of tumor), and irradiating tumor with a low-photon laser flux (1 W/cm(2)) to generate MHT. The treatment increased vascular permeability within 24 h after treatment, allowing enhanced transport of macromolecules up to 54 nm in size. A mathematical model is used to describe changes in tumor mass transport properties where the rate of macromolecular exchange between interstitial and vascular region (R) and maximum dye enhancement (Ymax) of 23-nm dextran dye is analytically solved. During enhanced permeability, R increased by 200% while Ymax increased by 30% relative to untreated group in pancreatic CAPAN-1 tumors. MHT treatment also enhanced transport of larger dextran dye (54 nm) as assessed by intravital microscopy, without causing occlusive cellular damage. Enhanced vascular transport was prolonged for up to 24 h after treatment, but reversible with transport parameters returning to basal levels after 36 h. This study indicates that localized mild hyperthermia treatment opens a transient time-window with which to enable and augment macromolecule transport and potentially improve therapeutic efficacy. From the clinical editor: In this study, local intra-tumor mild hyperthermia is induced using a nanoparticle-based approach utilizing stealth gold nanorods and irradiating the tumor with low-photon laser flux, resulting in locally increased vascular permeability enabling enhanced delivery of therapeutics, including macromolecules up to 54 nm in size. Similar approaches would be very helpful in addressing treatment-resistant malignancies in clinical practice.


Asunto(s)
Vasos Sanguíneos/metabolismo , Permeabilidad Capilar , Hipertermia Inducida , Neoplasias Pancreáticas/irrigación sanguínea , Animales , Transporte Biológico , Ratones , Ratones Desnudos , Nanotubos , Neoplasias Pancreáticas/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA