Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
medRxiv ; 2023 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-38234840

RESUMO

Glioblastoma (GBM) is a primary brain cancer with an abysmal prognosis and few effective therapies. The ability to investigate the tumor microenvironment before and during treatment would greatly enhance both understanding of disease response and progression, as well as the delivery and impact of therapeutics. Stereotactic biopsies are a routine surgical procedure performed primarily for diagnostic histopathologic purposes. The role of investigative biopsies - tissue sampling for the purpose of understanding tumor microenvironmental responses to treatment using integrated multi-modal molecular analyses ('Multi-omics") has yet to be defined. Secondly, it is unknown whether comparatively small tissue samples from brain biopsies can yield sufficient information with such methods. Here we adapt stereotactic needle core biopsy tissue in two separate patients. In the first patient with recurrent GBM we performed highly resolved multi-omics analysis methods including single cell RNA sequencing, spatial-transcriptomics, metabolomics, proteomics, phosphoproteomics, T-cell clonotype analysis, and MHC Class I immunopeptidomics from biopsy tissue that was obtained from a single procedure. In a second patient we analyzed multi-regional core biopsies to decipher spatial and genomic variance. We also investigated the utility of stereotactic biopsies as a method for generating patient derived xenograft models in a separate patient cohort. Dataset integration across modalities showed good correspondence between spatial modalities, highlighted immune cell associated metabolic pathways and revealed poor correlation between RNA expression and the tumor MHC Class I immunopeptidome. In conclusion, stereotactic needle biopsy cores are of sufficient quality to generate multi-omics data, provide data rich insight into a patient's disease process and tumor immune microenvironment and can be of value in evaluating treatment responses. One sentence summary: Integrative multi-omics analysis of stereotactic needle core biopsies in glioblastoma.

2.
Transl Oncol ; 21: 101427, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35472731

RESUMO

Long-term treatment outcomes for patients with high grade ovarian cancers have not changed despite innovations in therapies. There is no recommended assay for predicting patient response to second-line therapy, thus clinicians must make treatment decisions based on each individual patient. Patient-derived xenograft (PDX) tumors have been shown to predict drug sensitivity in ovarian cancer patients, but the time frame for intraperitoneal (IP) tumor generation, expansion, and drug screening is beyond that for tumor recurrence and platinum resistance to occur, thus results do not have clinical utility. We describe a drug sensitivity screening assay using a drug delivery microdevice implanted for 24 h in subcutaneous (SQ) ovarian PDX tumors to predict treatment outcomes in matched IP PDX tumors in a clinically relevant time frame. The SQ tumor response to local microdose drug exposure was found to be predictive of the growth of matched IP tumors after multi-week systemic therapy using significantly fewer animals (10 SQ vs 206 IP). Multiplexed immunofluorescence image analysis of phenotypic tumor response combined with a machine learning classifier could predict IP treatment outcomes against three second-line cytotoxic therapies with an average AUC of 0.91.

3.
Med Phys ; 46(11): 5134-5143, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31494942

RESUMO

PURPOSE: Recently developed implantable microdevices can perform multi-drug response assessment of cancer drugs in-vivo, with potential to develop highly optimized personalized cancer treatment strategies. However, minimally invasive/interventional image-guided methods of in-vivo microdevice implantation, securement, and retrieval are needed for broad clinical translation. Here we demonstrate proof-of-concept of an interventional microdevice implantation and retrieval method for personalized drug response assessment, using ex-vivo phantom, ex-vivo tissue, and in-vivo murine models. METHODS: A method for minimally-invasive microdevice implantation and retrieval was developed, by which a custom-prototyped 6 mm retrievable microdevice can be implanted into a live tumor, deliver drugs into 10 discrete regions of adjacent tissue, and retrieved along with the adjacent drug-exposed tissue with a custom-prototyped retrieval needle device to allow in-vivo multi-drug response assessment. Computed tomography (CT) and ultrasound (US)-guided minimally invasive microdevice implantation and retrieval were tested in ex-vivo phantom and tissue models. Successful retrieval was defined as retrieval of the microdevice and adjacent core phantom/tissue sample containing at least 4/10 drug delivery sites. Subsequently, 10 implantation and retrieval trials in phantom models were performed using bi-axial and tri-axial retrieval needles; success rates were calculated and compared using a two-proportion z-test and the number of successfully retrieved drug release sites per microdevice was calculated and compared using a one-tailed independent t-test. Finally, five microdevices, each containing ten reservoirs preloaded with chemotherapy agent Doxorubicin, were implanted into mouse tumors in-vivo, secured for 24-h during drug release, and microdevice/tissue retrieval was performed under ultrasound guidance. Fluorescence microscopy of the retrieved tissue was used to confirm drug delivery and apoptosis staining assessed in-vivo tissue response; correlation of drug release and apoptosis staining were used to assess in-vivo drug efficacy. RESULTS: Image-guided microdevice implantation and retrieval were successful in ex-vivo phantom and tissue models with both US and CT guidance. Bi-axial retrieval success rate was significantly higher than triaxial retrieval in ex-vivo phantom trials (90% vs 50%, z = 1.95, P = 0.026), and had nonsignificantly higher number of retrieved drug-release sites per microdevice (8.3 vs 7.0, t = 1.37, P = 0.097). Bi-axial retrieval was successful in all five in-vivo mouse tumor models, and allowed in-vivo drug response assessment at up to ten discrete drug delivery sites per microdevice. An average of 6.8/10 discrete tumor sites containing micro-doses of delivered drug were retrieved per in-vivo attempt (min 5, max 10, std 1.93). Tissue regions of drug delivery, as assessed with fluorescent Doxorubicin drug signal, correlated with regions of apoptosis staining in all in-vivo models, indicating drug efficacy. No bleeding, microdevice migration, or other complications were noted during implantation, 24-h observation, or retrieval. CONCLUSIONS: The demonstrated image-guided minimally invasive microdevice implantation and retrieval method is similar to routine outpatient biopsy procedures, obviates the need for surgery, and can be performed at varying depths under CT and/or US guidance. There is potential for this method to enable clinical translation of in-vivo personalized drug response assessment/prediction in a much larger number of patients than currently possible.


Assuntos
Microtecnologia/instrumentação , Imagens de Fantasmas , Próteses e Implantes , Cirurgia Assistida por Computador/instrumentação , Humanos , Medicina de Precisão , Resultado do Tratamento
4.
Small ; 15(37): e1901459, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31183933

RESUMO

Enhanced understanding of neuropathologies has created a need for more advanced tools. Current neural implants result in extensive glial scarring and are not able to highly localize drug delivery due to their size. Smaller implants reduce surgical trauma and improve spatial resolution, but such a reduction requires improvements in device design to enable accurate and chronic implantation in subcortical structures. Flexible needle steering techniques offer improved control over implant placement, but often require complex closed-loop control for accurate implantation. This study reports the development of steerable microinvasive neural implants (S-MINIs) constructed from borosilicate capillaries (OD = 60 µm, ID = 20 µm) that do not require closed-loop guidance or guide tubes. S-MINIs reduce glial scarring 3.5-fold compared to prior implants. Bevel steered needles are utilized for open-loop targeting of deep-brain structures. This study demonstrates a sinusoidal relationship between implant bevel angle and the trajectory radius of curvature both in vitro and ex vivo. This relationship allows for bevel-tipped capillaries to be steered to a target with an average error of 0.23 mm ± 0.19 without closed-loop control. Polished microcapillaries present a new microinvasive tool for chronic, predictable targeting of pathophysiological structures without the need for closed-loop feedback and complex imaging.


Assuntos
Procedimentos Cirúrgicos Robóticos/métodos , Animais , Encéfalo/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Desenho de Equipamento , Feminino , Humanos , Microscopia de Fluorescência/métodos , Imagens de Fantasmas , Ratos , Ratos Endogâmicos F344 , Suínos
5.
Analyst ; 143(20): 4836-4839, 2018 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-30070266

RESUMO

We describe a label-free approach based on Raman spectroscopy, to study drug-induced apoptosis in vivo. Spectral-shifts at wavenumbers associated with DNA, proteins, lipids, and collagen have been identified on breast and melanoma tumor tissues. These findings may enable a new analytical method for rapid readout of drug-therapy with miniaturized probes.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Melanoma/metabolismo , Análise Espectral Raman/métodos , Animais , Anticorpos/imunologia , Antineoplásicos/farmacologia , Caspase 3/imunologia , Caspase 3/metabolismo , Doxorrubicina/farmacologia , Imuno-Histoquímica , Substâncias Intercalantes/farmacologia , Camundongos Nus
6.
Nat Commun ; 9(1): 1184, 2018 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-29567939

RESUMO

The proliferation of computer-aided design and additive manufacturing enables on-demand fabrication of complex, three-dimensional structures. However, combining the versatility of cell-laden hydrogels within the 3D printing process remains a challenge. Herein, we describe a facile and versatile method that integrates polymer networks (including hydrogels) with 3D-printed mechanical supports to fabricate multicomponent (bio)materials. The approach exploits surface tension to coat fenestrated surfaces with suspended liquid films that can be transformed into solid films. The operating parameters for the process are determined using a physical model, and complex geometric structures are successfully fabricated. We engineer, by tailoring the window geometry, scaffolds with anisotropic mechanical properties that compress longitudinally (~30% strain) without damaging the hydrogel coating. Finally, the process is amenable to high cell density encapsulation and co-culture. Viability (>95%) was maintained 28 days after encapsulation. This general approach can generate biocompatible, macroscale devices with structural integrity and anisotropic mechanical properties.


Assuntos
Materiais Biocompatíveis/química , Fibroblastos/citologia , Hidrogéis/química , Impressão Tridimensional , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Desenho Assistido por Computador , Humanos , Impressão Tridimensional/instrumentação , Tensão Superficial , Engenharia Tecidual/instrumentação
7.
Gynecol Oncol ; 146(1): 161-169, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28499648

RESUMO

OBJECTIVE: Most ovarian cancer patients present with advanced-stage disease, disseminated in the peritoneal cavity. Standard treatment involves surgical resection of all visible tumor, followed by delivery of systemic therapy. Patients with advanced-stage disease may be candidates for intraperitoneal (IP) chemotherapy following surgical debulking. Recent clinical trials have created controversy regarding the benefits of this approach. Previous clinical trials report that patients with microscopic residual disease respond best to IP therapy. The goal of this study was to determine the relationship between tumor size and the efficacy of continuous chemotherapy. METHODS: Small and large ovarian cancer spheroids (derived from UCI101 and A2780 cell lines) were exposed to short-term high (modeling an IP injection, "IP") or prolonged, low cisplatin concentrations (modeling an implanted device, "device"), which have been previously shown to be less toxic. Spheroid diameter was measured at various time points via image analysis and used to quantify tumor shrinkage over the course of treatment. RESULTS: We show that "IP" doses more effectively shrink large spheroids when the same cumulative dose is administered with both treatments, but that both regimens similarly treat small spheroids. We also demonstrate that higher cumulative "device" doses are most effective at shrinking large spheroids. CONCLUSIONS: These results support the hypothesis that intratumoral drug distribution following IP treatment is diffusion-controlled. An implanted device that continuously delivers low doses of IP chemotherapy would, therefore, be maximally effective against microscopic tumors.


Assuntos
Antineoplásicos/administração & dosagem , Cisplatino/administração & dosagem , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Linhagem Celular Tumoral , Esquema de Medicação , Feminino , Humanos , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/patologia
8.
Nat Med ; 23(2): 235-241, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28024083

RESUMO

Mammalian tissues rely on a variety of nutrients to support their physiological functions. It is known that altered metabolism is involved in the pathogenesis of cancer, but which nutrients support the inappropriate growth of intact malignant tumors is incompletely understood. Amino acids are essential nutrients for many cancer cells that can be obtained through the scavenging and catabolism of extracellular protein via macropinocytosis. In particular, macropinocytosis can be a nutrient source for pancreatic cancer cells, but it is not fully understood how the tumor environment influences metabolic phenotypes and whether macropinocytosis supports the maintenance of amino acid levels within pancreatic tumors. Here we utilize miniaturized plasma exchange to deliver labeled albumin to tissues in live mice, and we demonstrate that breakdown of albumin contributes to the supply of free amino acids in pancreatic tumors. We also deliver albumin directly into tumors using an implantable microdevice, which was adapted and modified from ref. 9. Following implantation, we directly observe protein catabolism and macropinocytosis in situ by pancreatic cancer cells, but not by adjacent, non-cancerous pancreatic tissue. In addition, we find that intratumoral inhibition of macropinocytosis decreases amino acid levels. Taken together, these data suggest that pancreatic cancer cells consume extracellular protein, including albumin, and that this consumption serves as an important source of amino acids for pancreatic cancer cells in vivo.


Assuntos
Aminoácidos/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/metabolismo , Pinocitose , Proteólise , Albumina Sérica/metabolismo , Albuminas/metabolismo , Animais , Linhagem Celular Tumoral , Cromatografia Gasosa , Modelos Animais de Doenças , Espaço Extracelular/metabolismo , Camundongos , Microscopia de Fluorescência por Excitação Multifotônica , Isótopos de Nitrogênio , Plasmaferese , Proteínas/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
9.
Nat Commun ; 7: 11987, 2016 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-27329820

RESUMO

Identifying therapeutic targets in rare cancers remains challenging due to the paucity of established models to perform preclinical studies. As a proof-of-concept, we developed a patient-derived cancer cell line, CLF-PED-015-T, from a paediatric patient with a rare undifferentiated sarcoma. Here, we confirm that this cell line recapitulates the histology and harbours the majority of the somatic genetic alterations found in a metastatic lesion isolated at first relapse. We then perform pooled CRISPR-Cas9 and RNAi loss-of-function screens and a small-molecule screen focused on druggable cancer targets. Integrating these three complementary and orthogonal methods, we identify CDK4 and XPO1 as potential therapeutic targets in this cancer, which has no known alterations in these genes. These observations establish an approach that integrates new patient-derived models, functional genomics and chemical screens to facilitate the discovery of targets in rare cancers.


Assuntos
Quinase 4 Dependente de Ciclina/genética , Carioferinas/genética , Doenças Raras/genética , Receptores Citoplasmáticos e Nucleares/genética , Sarcoma/genética , Células A549 , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Sistemas CRISPR-Cas , Ciclo Celular , Linhagem Celular Tumoral , Doxorrubicina/administração & dosagem , Ensaios de Seleção de Medicamentos Antitumorais , Exoma , Feminino , Genômica , Humanos , Hidrazinas/administração & dosagem , Camundongos , Camundongos Nus , Metástase Neoplásica , Recidiva Local de Neoplasia , Transplante de Neoplasias , Piperazinas/administração & dosagem , Piridinas/administração & dosagem , Interferência de RNA , Doenças Raras/tratamento farmacológico , Sarcoma/tratamento farmacológico , Análise de Sequência de RNA , Triazóis/administração & dosagem , Proteína Exportina 1
10.
Clin Cancer Res ; 22(24): 6031-6038, 2016 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-27091406

RESUMO

PURPOSE: Treatment of BRAF-mutated melanoma tumors with BRAF inhibitor-based therapy produces high response rates, but of limited duration in the vast majority of patients. Published investigations of resistance mechanisms suggest numerous examples of tumor adaptation and signal transduction bypass mechanisms, but without insight into biomarkers that would predict which mechanism will predominate. Monitoring phenotypic response of multiple adaptive mechanisms simultaneously within the same tumor as it adapts during treatment has been elusive. EXPERIMENTAL DESIGN: This study reports on a method to provide a more complete understanding of adaptive tumor responses. We simultaneously measured in vivo antitumor activity of 12 classes of inhibitors, which are suspected of enabling adaptive escape mechanisms, at various time points during systemic BRAF inhibition. We used implantable microdevices to release multiple compounds into distinct regions of a tumor to measure the efficacy of each compound independently and repeated these measurements as tumors progressed on systemic BRAF treatment. RESULTS: We observed varying phenotypic responses to specific inhibitors before, during, and after prolonged systemic treatment with BRAF inhibitors. Our results specifically identify PI3K, PDGFR, EGFR, and HDAC inhibitors as becoming significantly more efficacious during systemic BRAF inhibition. The sensitivity to other targeted inhibitors remained mostly unchanged, whereas local incremental sensitivity to PLX4720 declined sharply. CONCLUSIONS: These findings suggest redundancy of several resistance mechanisms and may help identify optimal constituents of more effective combination therapy in BRAF-mutant melanoma. They also represent a new paradigm for dynamic measurement of adaptive signaling mechanisms within the same tumor during therapy. Clin Cancer Res; 22(24); 6031-8. ©2016 AACR.


Assuntos
Antineoplásicos/farmacologia , Melanoma/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Humanos , Indóis/metabolismo , Melanoma/metabolismo , Camundongos , Camundongos Nus , Mutação/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
11.
Cancer Discov ; 6(5): 516-31, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26811325

RESUMO

UNLABELLED: Fibronectin (FN) is a major component of the tumor microenvironment, but its role in promoting metastasis is incompletely understood. Here, we show that FN gradients elicit directional movement of breast cancer cells, in vitro and in vivo Haptotaxis on FN gradients requires direct interaction between α5ß1 integrin and MENA, an actin regulator, and involves increases in focal complex signaling and tumor cell-mediated extracellular matrix (ECM) remodeling. Compared with MENA, higher levels of the prometastatic MENA(INV) isoform associate with α5, which enables 3-D haptotaxis of tumor cells toward the high FN concentrations typically present in perivascular space and in the periphery of breast tumor tissue. MENA(INV) and FN levels were correlated in two breast cancer cohorts, and high levels of MENA(INV) were significantly associated with increased tumor recurrence as well as decreased patient survival. Our results identify a novel tumor cell-intrinsic mechanism that promotes metastasis through ECM remodeling and ECM-guided directional migration. SIGNIFICANCE: Here, we provide new insight into how tumor cell:ECM interactions generate signals and structures that promote directed tumor cell migration, a critical component of metastasis. Our results identify a tumor cell-intrinsic mechanism driven by the actin regulatory protein MENA that promotes ECM remodeling and haptotaxis along FN gradients. Cancer Discov; 6(5); 516-31. ©2016 AACR.See related commentary by Santiago-Medina and Yang, p. 474This article is highlighted in the In This Issue feature, p. 461.


Assuntos
Movimento Celular , Matriz Extracelular/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Actinas/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Colágeno/genética , Colágeno/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Matriz Extracelular/genética , Feminino , Fibronectinas/genética , Fibronectinas/metabolismo , Expressão Gênica , Xenoenxertos , Humanos , Integrina alfa5beta1/metabolismo , Estimativa de Kaplan-Meier , Camundongos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Metástase Neoplásica , Neoplasias/genética , Neoplasias/mortalidade , Prognóstico , Ligação Proteica , Transdução de Sinais , Microambiente Tumoral
12.
J Control Release ; 220(Pt A): 358-367, 2015 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-26548976

RESUMO

Intraperitoneal (IP) chemotherapy for ovarian cancer treatment prolongs overall survival by 16 months compared to intravenous chemotherapy but is not widely practiced due to catheter-related complications and complexity of administration. An implantable, nonresorbable IP microdevice was used to release chemotherapeutic agent at a constant rate of approximately 1.3 µg/h in vitro and 1.0 µg/h in vivo. Studies conducted in two orthotopic murine models bearing human xenografts (SKOV3 and UCI101) demonstrate that continuous dosing reduces tumor burden to the same extent as weekly IP bolus drug injections. Treatment-induced toxicity was quantified via body weight loss and complete blood count. The microdevice resulted in significantly less toxicity than IP bolus injections, despite administration of higher cumulative doses (total area under the concentration-time curve of 3049 ng day/mL with the microdevice vs. 2118 ng-day/mL with IP bolus injections). This preclinical study supports the concept that reduced toxicity with similar efficacy outcomes can be achieved by continuous dosing in ovarian cancer patients currently treated with IP therapy.


Assuntos
Antineoplásicos/administração & dosagem , Cisplatino/administração & dosagem , Neoplasias Ovarianas/tratamento farmacológico , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidade , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/química , Cisplatino/farmacocinética , Cisplatino/toxicidade , Composição de Medicamentos , Implantes de Medicamento , Feminino , Humanos , Injeções Intravenosas , Leucopenia/induzido quimicamente , Camundongos Nus , Miniaturização , Neoplasias Ovarianas/patologia , Solubilidade , Carga Tumoral/efeitos dos fármacos , Redução de Peso/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
13.
J Control Release ; 217: 183-90, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26285064

RESUMO

Treatments of brain tumor associated edema with systemically delivered dexamethasone, the standard of care, and cediranib, a novel anti-edema agent, are associated with systemic toxicities in brain tumor patients. A tunable, reservoir-based drug delivery device was developed to investigate the effects of delivering dexamethasone and cediranib locally in the brain in an intracranial 9L gliosarcoma rat model. Reproducible, sustained releases of both dexamethasone and solid dispersion of cediranib in polyvinylpyrrolidone (AZD/PVP) from these devices were achieved. The water-soluble AZD/PVP, which exhibited similar bioactivity as cediranib, was developed to enhance the release of cediranib from the device. Local and systemic administration of both dexamethasone and cediranib was equally efficacious in alleviating edema but had no effect on tumor growth. Edema reduction led to modest but significant improvement in survival. Local delivery of dexamethasone prevented dexamethasone-induced weight loss, an adverse effect seen in animals treated with systemic dexamethasone. Local deliveries of dexamethasone and cediranib via these devices used only 2.36% and 0.21% of the systemic doses respectively, but achieved similar efficacy as systemic drug deliveries without the side effects associated with systemic administration. Other therapeutic agents targeting brain tumor can be delivered locally in the brain to provide similar improved treatment outcomes.


Assuntos
Anti-Inflamatórios/administração & dosagem , Antineoplásicos/administração & dosagem , Dexametasona/administração & dosagem , Sistemas de Liberação de Medicamentos , Edema/tratamento farmacológico , Quinazolinas/administração & dosagem , Animais , Anti-Inflamatórios/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/complicações , Neoplasias Encefálicas/tratamento farmacológico , Células Cultivadas , Dexametasona/uso terapêutico , Liberação Controlada de Fármacos , Edema/etiologia , Feminino , Glioma/complicações , Glioma/tratamento farmacológico , Células Endoteliais da Veia Umbilical Humana , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/uso terapêutico , Quinazolinas/uso terapêutico , Ratos Endogâmicos F344
14.
PLoS One ; 10(5): e0126855, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26000958

RESUMO

Atherosclerosis and malignancy are pervasive pathological conditions that account for the bulk of morbidity and mortality in developed countries. Our current understanding of the patholobiology of these fundamental disorders suggests that inflammatory processes may differentially affect them; thus, atherosclerosis can be largely driven by inflammation, where as cancer often flourishes as inflammatory responses are modulated. A corollary of this hypothesis is that cancer (or its treatment may significantly attenuate atherosclerotic disease by diminishing host inflammatory response, suggesting potential therapeutic approaches. To evaluate the relationship between cancer and cardiovascular atherosclerotic disease, we assessed 1,024 autopsy reports from Brigham and Women's Hospital and performed correlative analyses on atherosclerotic severity and cancer prevalence. In gender- and age-matched populations, there is a statistically significant inverse correlation between history of malignancy and autopsy-proven atherosclerotic disease. In a second analysis, we evaluated 147,779 patients through analysis of the Harvard Catalyst SHRINE database and demonstrated a reduced non-coronary atherosclerotic disease rate: control (27.40%), leukemia/lymphoma (12.57%), lung (17.63%), colorectal (18.17%), breast (9.79%), uterus/cervix (11.47%), and prostate (18.40%). We herein report that, based on two separate medical records analysis, an inverse correlation between cancer and atherosclerosis. Furthermore, this correlation is not uniformly associated with anti-neoplastic treatment, suggesting that the inverse relationship may be in part attributable to an individual's intrinsic inflammatory propensity, and/or to inflammation-modulatory properties of neoplasms.


Assuntos
Aterosclerose/epidemiologia , Neoplasias/epidemiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Aterosclerose/diagnóstico , Feminino , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Prevalência , Risco , Índice de Gravidade de Doença , Adulto Jovem
15.
Sci Transl Med ; 7(284): 284ra57, 2015 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-25904741

RESUMO

Current anticancer chemotherapy relies on a limited set of in vitro or indirect prognostic markers of tumor response to available drugs. A more accurate analysis of drug sensitivity would involve studying tumor response in vivo. To this end, we have developed an implantable device that can perform drug sensitivity testing of several anticancer agents simultaneously inside the living tumor. The device contained reservoirs that released microdoses of single agents or drug combinations into spatially distinct regions of the tumor. The local drug concentrations were chosen to be representative of concentrations achieved during systemic treatment. Local efficacy and drug concentration profiles were evaluated for each drug or drug combination on the device, and the local efficacy was confirmed to be a predictor of systemic efficacy in vivo for multiple drugs and tumor models. Currently, up to 16 individual drugs or combinations can be assessed independently, without systemic drug exposure, through minimally invasive biopsy of a small region of a single tumor. This assay takes into consideration physiologic effects that contribute to drug response by allowing drugs to interact with the living tumor in its native microenvironment. Because these effects are crucial to predicting drug response, we envision that these devices will help identify optimal drug therapy before systemic treatment is initiated and could improve drug response prediction beyond the biomarkers and in vitro and ex vivo studies used today. These devices may also be used in clinical drug development to safely gather efficacy data on new compounds before pharmacological optimization.


Assuntos
Antineoplásicos/uso terapêutico , Monitoramento de Medicamentos/instrumentação , Ensaios de Seleção de Medicamentos Antitumorais/instrumentação , Neoplasias/tratamento farmacológico , Animais , Apoptose , Biomarcadores Tumorais , Biópsia , Calibragem , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Doxorrubicina/química , Sistemas de Liberação de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Humanos , Camundongos , Polietilenoglicóis/química , Polímeros/química , Prognóstico
16.
Mol Ther ; 22(5): 999-1007, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24496384

RESUMO

The secreted proteins from a cell constitute a natural biologic library that can offer significant insight into human health and disease. Discovering new secreted proteins from cells is bounded by the limitations of traditional separation and detection tools to physically fractionate and analyze samples. Here, we present a new method to systematically identify bioactive cell-secreted proteins that circumvent traditional proteomic methods by first enriching for protein candidates by differential gene expression profiling. The bone marrow stromal cell secretome was analyzed using enriched gene expression datasets in combination with potency assay testing. Four proteins expressed by stromal cells with previously unknown anti-inflammatory properties were identified, two of which provided a significant survival benefit to mice challenged with lethal endotoxic shock. Greater than 85% of secreted factors were recaptured that were otherwise undetected by proteomic methods, and remarkable hit rates of 18% in vitro and 9% in vivo were achieved.


Assuntos
Proteínas Contráteis/genética , Proteínas Contráteis/metabolismo , Encefalinas/genética , Proteínas da Matriz Extracelular/metabolismo , Glicoproteínas/genética , Interleucina-10/metabolismo , Precursores de Proteínas/genética , Proteínas/metabolismo , Choque Séptico/terapia , Animais , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Encefalinas/metabolismo , Proteínas da Matriz Extracelular/genética , Perfilação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Interleucina-10/genética , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Biossíntese de Proteínas/genética , Precursores de Proteínas/metabolismo , Proteínas/genética , Proteômica , Fatores de Processamento de RNA , Choque Séptico/genética
17.
Biomaterials ; 33(23): 5768-75, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22591609

RESUMO

Primary malignant brain tumors (BT) are the most common and aggressive malignant brain tumor. Treatment of BTs is a daunting task with median survival just at 21 months. Methods of localized delivery have achieved success in treating BT by circumventing the blood brain barrier and achieving high concentrations of therapeutic within the tumor. The capabilities of localized delivery can be enhanced by utilizing mirco-electro-mechanical systems (MEMS) technology to deliver drugs with precise temporal control over release kinetics. An intracranial MEMS based device was developed to deliver the clinically utilized chemotherapeutic temozolomide (TMZ) in a rodent glioma model. The device is a liquid crystalline polymer reservoir, capped by a MEMS microchip. The microchip contains three nitride membranes that can be independently ruptured at any point during or after implantation. The kinetics of TMZ release were validated and quantified in vitro. The safety of implanting the device intracranially was confirmed with preliminary in vivo studies. The impact of TMZ release kinetics was investigated by conducting in vivo studies that compared the effects of drug release rates and timing on animal survival. TMZ delivered from the device was effective at prolonging animal survival in a 9L rodent glioma model. Immunohistological analysis confirmed that TMZ was released in a viable, cytotoxic form. The results from the in vivo efficacy studies indicate that early, rapid delivery of TMZ from the device results in the most prolonged animal survival. The ability to actively control the rate and timing of drug(s) release holds tremendous potential for the treatment of BTs and related diseases.


Assuntos
Antineoplásicos Alquilantes/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/análogos & derivados , Sistemas de Liberação de Medicamentos/instrumentação , Gliossarcoma/tratamento farmacológico , Animais , Antineoplásicos Alquilantes/farmacocinética , Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/patologia , Dacarbazina/administração & dosagem , Dacarbazina/farmacocinética , Dacarbazina/uso terapêutico , Desenho de Equipamento , Feminino , Gliossarcoma/patologia , Ratos , Ratos Endogâmicos F344 , Temozolomida
18.
Biomaterials ; 32(10): 2532-9, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21220172

RESUMO

Controlled-release drug delivery systems are capable of treating debilitating diseases, including cancer. Brain cancer, in particular glioblastoma multiforme (GBM), is an extremely invasive cancer with a dismal prognosis. The use of drugs capable of crossing the blood-brain barrier has shown modest prolongation in patient survival, but not without unsatisfactory systemic, dose-limiting toxicity. Among the reasons for this improvement include a better understanding of the challenges of delivery of effective agents directly to the brain tumor site. The combination of carmustine delivered by biodegradable polyanhydride wafers (Gliadel(®)), with the systemic alkylating agent, temozolomide, allows much higher effective doses of the drug while minimizing the systemic toxicity. We have previously shown that locally delivering these two drugs leads to further improvement in survival in experimental models. We postulated that microcapsule devices capable of releasing temozolomide would increase the therapeutic capability of this approach. A biocompatible drug delivery microcapsule device for the intracranial delivery of temozolomide is described. Drug release profiles from these microcapsules can be modulated based on the physical chemistry of the drug and the dimensions of the release orifices in these devices. The drug released from the microcapsules in these experiments was the clinically utilized chemotherapeutic agent, temozolomide. In vitro studies were performed in order to test the function, reliability, and drug release kinetics of the devices. The efficacy of the temozolomide-filled microcapsules was tested in an intracranial experimental rodent gliosarcoma model. Immunohistochemical analysis of tissue for evidence of DNA strand breaks via terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay was performed. The experimental release curves showed mass flow rates of 36 µg/h for single-orifice devices and an 88 µg/h mass flow rate for multiple-orifice devices loaded with temozolomide. In vivo efficacy results showed that localized intracranial delivery of temozolomide from microcapsule devices was capable of prolonging animal survival and may offer a novel form of treatment for brain tumors.


Assuntos
Neoplasias Encefálicas/terapia , Encéfalo/patologia , Cápsulas/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Gliossarcoma/terapia , Animais , Encéfalo/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Dacarbazina/administração & dosagem , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Modelos Animais de Doenças , Gliossarcoma/patologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Cinética , Ratos , Ratos Endogâmicos F344 , Temozolomida
19.
Biosens Bioelectron ; 24(11): 3252-7, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19442510

RESUMO

Biopsies provide required information to diagnose cancer but, because of their invasiveness, they are difficult to use for managing cancer therapy. The ability to repeatedly sample the local environment for tumor biomarker, chemotherapeutic agent, and tumor metabolite concentrations could improve early detection of metastasis and personalized therapy. Here we describe an implantable diagnostic device that senses the local in vivo environment. This device, which could be left behind during biopsy, uses a semi-permeable membrane to contain nanoparticle magnetic relaxation switches. A cell line secreting a model cancer biomarker produced ectopic tumors in mice. The transverse relaxation time (T(2)) of devices in tumor-bearing mice was 20+/-10% lower than devices in control mice after 1 day by magnetic resonance imaging (p<0.01). Short term applications for this device are numerous, including verification of successful tumor resection. This may represent the first continuous monitoring device for soluble cancer biomarkers in vivo.


Assuntos
Biomarcadores Tumorais/análise , Técnicas Biossensoriais/instrumentação , Coriocarcinoma/imunologia , Coriocarcinoma/mortalidade , Imunoensaio/instrumentação , Magnetismo/instrumentação , Monitorização Ambulatorial/instrumentação , Próteses e Implantes , Animais , Biomarcadores Tumorais/imunologia , Linhagem Celular Tumoral , Coriocarcinoma/patologia , Desenho de Equipamento , Análise de Falha de Equipamento , Feminino , Camundongos , Camundongos Nus , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA