Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Gastroenterol ; 110(7): 948-55, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25601013

RESUMEN

OBJECTIVES: As there are no US population-based studies examining Lynch syndrome (LS) screening frequency by microsatellite instability (MSI) and immunohistochemistry (IHC), we seek to quantitate statewide rates in patients aged ≤50 years using data from a Centers for Disease Control and Prevention-funded Comparative Effectiveness Research (CER) project and identify factors associated with testing. Screening rates in this young, high-risk population may provide a best-case scenario as older patients, potentially deemed lower risk, may undergo testing less frequently. We also seek to determine how frequently MSI/IHC results are available preoperatively, as this may assist with decisions regarding colonic resection extent. METHODS: Data from all Louisiana colorectal cancer (CRC) patients aged ≤50 years diagnosed in 2011 were obtained from the Louisiana Tumor Registry CER project. Registry researchers and physicians analyzed data, including pathology and MSI/IHC. RESULTS: Of the 2,427 statewide all-age CRC patients, there were 274 patients aged ≤50 years, representing health care at 61 distinct facilities. MSI and/or IHC were performed in 23.0% of patients. Testing-associated factors included CRC family history (P<0.0045), urban location (P<0.0370), and care at comprehensive cancer centers (P<0.0020) but not synchronous/metachronous CRC or MSI-like histology. Public hospital screening was disproportionately low (P<0.0217). Of those tested, MSI and/or IHC was abnormal in 21.7%. Of those with abnormal IHC, staining patterns were consistent with LS in 87.5%. MSI/IHC results were available preoperatively in 16.9% of cases. CONCLUSIONS: Despite frequently abnormal MSI/IHC results, LS screening in young, high-risk patients is low. Provider education and disparities in access to specialized services, particularly in underserved populations, are possible contributors. MSI/IHC results are infrequently available preoperatively.


Asunto(s)
Colectomía , Neoplasias Colorrectales Hereditarias sin Poliposis/diagnóstico , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Detección Precoz del Cáncer , Pruebas Genéticas , Tamizaje Masivo , Inestabilidad de Microsatélites , Adulto , Factores de Edad , Neoplasias Colorrectales Hereditarias sin Poliposis/epidemiología , Neoplasias Colorrectales Hereditarias sin Poliposis/prevención & control , Neoplasias Colorrectales Hereditarias sin Poliposis/cirugía , Factores de Confusión Epidemiológicos , Detección Precoz del Cáncer/métodos , Detección Precoz del Cáncer/normas , Detección Precoz del Cáncer/estadística & datos numéricos , Femenino , Humanos , Inmunohistoquímica , Louisiana/epidemiología , Masculino , Tamizaje Masivo/métodos , Tamizaje Masivo/normas , Tamizaje Masivo/estadística & datos numéricos , Persona de Mediana Edad , Neoplasias Primarias Múltiples/genética , Neoplasias Primarias Secundarias/genética , Periodo Preoperatorio , Prevalencia , Población Rural/estadística & datos numéricos , Factores de Tiempo , Estados Unidos/epidemiología , Población Urbana/estadística & datos numéricos
2.
Cancer ; 120 Suppl 23: 3793-806, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25412391

RESUMEN

BACKGROUND: The Collaborative Stage (CS) Data Collection System enables multiple cancer registration programs to document anatomic and molecular pathology features that contribute to the Tumor (T), Node (N), Metastasis (M) - TNM - system of the American Joint Committee on Cancer (AJCC). This article highlights changes in CS for colon and rectal carcinomas as TNM moved from the AJCC 6th to the 7th editions. METHODS: Data from 18 Surveillance, Epidemiology, and End Results (SEER) population-based registries were analyzed for the years 2004-2010, which included 191,361colon and 73,341 rectal carcinomas. RESULTS: Overall, the incidence of colon and rectal cancers declined, with the greatest decrease in stage 0. The AJCC's 7th edition introduction of changes in the subcategorization of T4, N1, and N2 caused shifting within stage groups in 25,577 colon and 10,150 rectal cancers diagnosed in 2010. Several site-specific factors (SSFs) introduced in the 7th edition had interesting findings: 1) approximately 10% of colon and rectal cancers had tumor deposits - about 30%-40% occurred without lymph node metastases, which resulted in 2.5% of colon and 3.3% of rectal cases becoming N1c (stage III A/B) in the AJCC 7th edition; 2) 10% of colon and 12% of rectal cases had circumferential radial margins <1 mm; 3) about 46% of colorectal cases did not have a carcinoembryonic antigen (CEA) testing or documented CEA information; and 4) about 10% of colorectal cases had perineural invasion. CONCLUSIONS: Adoption of the AJCC 7th edition by the SEER program provides an assessment tool for staging and SSFs on clinical outcomes. This evidence can be used for education and improved treatment for colorectal carcinomas.


Asunto(s)
Carcinoma/patología , Neoplasias del Colon/patología , Ganglios Linfáticos/patología , Neoplasias del Recto/patología , Sistema de Registros , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Antígeno Carcinoembrionario/sangre , Carcinoma/metabolismo , Estudios de Cohortes , Neoplasias del Colon/metabolismo , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Humanos , Estadificación de Neoplasias/tendencias , Neoplasias del Recto/metabolismo , Estudios Retrospectivos , Programa de VERF
3.
J Pathol ; 221(4): 361-2, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20593484

RESUMEN

This Invited Response addresses concerns and opinions expressed in an Invited Commentary, 'Evidence-based medicine: the time has come to set standards for staging', by Quirke et al., published in this issue of The Journal of Pathology.


Asunto(s)
Estadificación de Neoplasias/normas , Neoplasias/patología , Neoplasias Colorrectales/patología , Medicina Basada en la Evidencia/métodos , Humanos , Neoplasias Pulmonares/patología , Metástasis Linfática , Estadificación de Neoplasias/métodos
4.
Hum Vaccin Immunother ; 16(3): 636-644, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-31584324

RESUMEN

Treatments with cytotoxic agents or viruses may cause Immunogenic Cell Death (ICD) that immunize tumor-bearing hosts but do not cause complete regression of tumor. We postulate that combining two ICD inducers may cause durable regression in immunocompetent mice. ICD was optimized in vitro by maximizing calreticulin externalization in human colorectal carcinoma (CRC) cells by exposure to mixtures of Oxaliplatin (OX) and human adenovirus (AdV). Six mm diameter CT26 or 4T1 carcinomas in flanks of BALB/c mice were injected once intratumorally (IT) with OX, AdV or their mixture. Tumor growth, Tumor-Infiltrating Lymphocytes (TIL), nodal cytotoxicity, and rejection of a viable cell challenge were measured. Tumors injected IT once with an optimum mixture of 80 µM OX - AdV 25 Multiplicity of Infection (MOI) in PBS buffer were 17-29% the volume of control tumors. When buffer was changed from PBS to 5% dextrose in water (D5W), volumes of tumors injected IT with 80 µM OX-AdV 25 MOI were 10% while IT OX or AdV alone were 32% and 40% the volume of IT buffer-treated tumors. OX-AdV IT increased CD3+ TIL by 4-fold, decreased CD8+ PD-1+ TIL from 79% to 19% and induced cytotoxicity to CT26 cells in draining node lymphocytes while lymphocytes from CT26-bearing untreated mice were not cytotoxic. OX-AdV IT in D5W caused complete regression in 40% of mice. Long-term survivors rejected a contralateral challenge of CT26. The buffer for Oxaliplatin is critical. The two ICD inducer mixture is promising as an agnostic sensitizer for carcinomas like colorectal carcinoma.


Asunto(s)
Neoplasias Colorrectales , Muerte Celular Inmunogénica , Adenoviridae/genética , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Oxaliplatino
5.
Cancer Res ; 67(10): 4774-82, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17510406

RESUMEN

Carcinoembryonic antigen (CEA) is a tumor marker that is associated with metastasis, poor response to chemotherapy of colorectal cancer (CRC), and anoikis, a form of apoptosis caused by cell detachment from matrix that is dependent on TRAIL-R2 (DR5) and caspase-8 activation in CRC. Although CEA is a homophilic binding protein that may provide survival signals through homotypical cell aggregation, we now report that CEA binds TRAIL-R2 (DR5) directly in two-hybrid assays to decrease anoikis through the extrinsic pathway. Deletion of the PELPK sequence (delPELPK) of CEA (delPELPK CEA) restores sensitivity to anoikis while it maintains its cell aggregation function. Wild-type (WT) CEA also increases experimental hepatic metastasis, whereas the delPELPK CEA does not. Thus, membrane CEA interacts with DR5 to inhibit anoikis and increase metastatic potential in CRC.


Asunto(s)
Anoicis/fisiología , Antígeno Carcinoembrionario/metabolismo , Neoplasias Colorrectales/patología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Antígeno Carcinoembrionario/biosíntesis , Antígeno Carcinoembrionario/genética , Caspasa 8/metabolismo , Inhibidores de Caspasas , Adhesión Celular/fisiología , Neoplasias Colorrectales/metabolismo , Células HT29 , Humanos , Integrinas/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Unión Proteica , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/antagonistas & inhibidores , Transducción de Señal/fisiología
6.
Mol Cancer Res ; 17(1): 42-53, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30201826

RESUMEN

MCL-1, a member of the antiapoptotic BCL-2 family, is a prosurvival protein with an essential DNA repair function. This study aims to test whether inhibition of protein synthesis by mTOR complex (mTORC) inhibitors depletes MCL-1, suppresses homologous recombination (HR) repair, and sensitizes cancer cells to PARP inhibitors. Treatment with everolimus decreases MCL-1 in colorectal carcinomas and small cell lung cancer (SCLC) cells but not glioblastoma multiforme (GBM) cells with a PTEN mutational background. However, AZD2014, a dual mTORC inhibitor, depletes MCL-1 in GBMs. Further, we show that everolimus decreases 4EBP1 phosphorylation only in colorectal carcinoma, whereas AZD2014 decreases 4EBP1 phosphorylation in both colorectal carcinoma and GBM cells. Combination therapy using everolimus or AZD2014 with olaparib inhibits the growth of clone A and U87-MG xenografts in in vivo and decreases clonogenic survival in in vitro compared with monotherapy. Reintroduction of MCL-1 rescues the survival of cancer cells in response to combination of everolimus or AZD2014 with olaparib. Treatment of cells with mTORC inhibitors and olaparib increases γ-H2AX and 53BP1 foci, decreases BRCA1, RPA, and Rad51 foci, impairs phosphorylation of ATR/Chk1 kinases, and induces necroptosis. In summary, mTORC inhibitors deplete MCL-1 to suppress HR repair and increase sensitivity to olaparib both in in vitro and in xenografts. IMPLICATIONS: Targeting the DNA repair activity of MCL-1 in in vivo for cancer therapy has not been tested. This study demonstrates that depleting MCL-1 sensitizes cancer cells to PARP inhibitors besides eliciting necroptosis, which could stimulate antitumor immunity to improve the therapeutic intervention of cancers.


Asunto(s)
Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Neoplasias/tratamiento farmacológico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Benzamidas/administración & dosificación , Benzamidas/farmacología , Línea Celular Tumoral , Daño del ADN , Reposicionamiento de Medicamentos , Sinergismo Farmacológico , Everolimus/administración & dosificación , Everolimus/farmacología , Femenino , Humanos , Masculino , Ratones , Ratones Desnudos , Morfolinas/administración & dosificación , Morfolinas/farmacología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Neoplasias/genética , Neoplasias/metabolismo , Ftalazinas/administración & dosificación , Ftalazinas/farmacología , Piperazinas/administración & dosificación , Piperazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/administración & dosificación , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cytometry B Clin Cytom ; 94(2): 239-249, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28475275

RESUMEN

BACKGROUND: Minimal residual disease (MRD) in B lymphoblastic leukemia (B-ALL) by flow cytometry is an established prognostic factor used to adjust treatment in most pediatric therapeutic protocols. MRD in B-ALL has been standardized by the Children's Oncology Group (COG) in North America, but not routine clinical labs. The Foundation for National Institutes of Health sought to harmonize MRD measurement among COG, oncology groups, academic, community and government, laboratories. METHODS: Listmode data from post-induction marrows were distributed from a reference lab to seven different clinical FCM labs with variable experience in B-ALL MRD. Labs were provided with the COG protocol. Files from 15 cases were distributed to the seven labs. Educational sessions were implemented, and 10 more listmode file cases analyzed. RESULTS: Among 105 initial challenges, the overall discordance rate was 26%. In the final round, performance improved considerably; out of 70 challenges, there were five false positives and one false negative (9% discordance), and no quantitative discordance. Four of six deviations occurred in a single lab. Three samples with hematogones were still misclassified as MRD. CONCLUSIONS: Despite the provision of the COG standardized analysis protocol, even experienced laboratories require an educational component for B-ALL MRD analysis by FCM. Recognition of hematogones remains challenging for some labs when using the COG protocol. The results from this study suggest that dissemination of MRD testing to other North American laboratories as part of routine clinical management of B-ALL is possible but requires additional educational components to complement standardized methodology. © 2017 International Clinical Cytometry Society.


Asunto(s)
Neoplasia Residual/diagnóstico , Neoplasia Residual/patología , Citometría de Flujo/métodos , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Pronóstico
8.
JAMA Oncol ; 3(7): e170580, 2017 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-28494052

RESUMEN

IMPORTANCE: Minimal residual disease (MRD) refers to the presence of disease in cases deemed to be in complete remission by conventional pathologic analysis. Assessing the association of MRD status following induction therapy in patients with acute lymphoblastic leukemia (ALL) with relapse and mortality may improve the efficiency of clinical trials and accelerate drug development. OBJECTIVE: To quantify the relationships between event-free survival (EFS) and overall survival (OS) with MRD status in pediatric and adult ALL using publications of clinical trials and other databases. DATA SOURCES: Clinical studies in ALL identified via searches of PubMed, MEDLINE, and clinicaltrials.gov. STUDY SELECTION: Our search and study screening process adhered to the PRISMA Guidelines. Studies that addressed EFS or OS by MRD status in patients with ALL were included; reviews, abstracts, and studies with fewer than 30 patients or insufficient MRD description were excluded. DATA EXTRACTION AND SYNTHESIS: Study sample size, patient age, follow-up time, timing of MRD assessment (postinduction or consolidation), MRD detection method, phenotype/genotype (B cell, T cell, Philadelphia chromosome), and EFS and OS. Searches of PubMed and MEDLINE identified 566 articles. A parallel search on clinicaltrials.gov found 67 closed trials and 62 open trials as of 2014. Merging results of 2 independent searches and applying exclusions gave 39 publications in 3 arms of patient populations (adult, pediatric, and mixed). We performed separate meta-analyses for each of these 3 subpopulations. RESULTS: The 39 publications comprised 13 637 patients: 16 adult studies (2076 patients), 20 pediatric (11 249 patients), and 3 mixed (312 patients). The EFS hazard ratio (HR) for achieving MRD negativity is 0.23 (95% Bayesian credible interval [BCI] 0.18-0.28) for pediatric patients and 0.28 (95% BCI, 0.24-0.33) for adults. The respective HRs in OS are 0.28 (95% BCI, 0.19-0.41) and 0.28 (95% BCI, 0.20-0.39). The effect was similar across all subgroups and covariates. CONCLUSIONS AND RELEVANCE: The value of having achieved MRD negativity is substantial in both pediatric and adult patients with ALL. These results are consistent across therapies, methods of and times of MRD assessment, cutoff levels, and disease subtypes. Minimal residual disease status warrants consideration as an early measure of disease response for evaluating new therapies, improving the efficiency of clinical trials, accelerating drug development, and for regulatory approval. A caveat is that an accelerated approval of a particular new drug using an intermediate end point, such as MRD, would require confirmation using traditional efficacy end points.


Asunto(s)
Neoplasia Residual/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Adulto , Niño , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Pronóstico , Análisis de Supervivencia , Resultado del Tratamiento
9.
Clin Cancer Res ; 23(15): 3980-3993, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28428191

RESUMEN

Treatment of myeloma has benefited from the introduction of more effective and better tolerated agents, improvements in supportive care, better understanding of disease biology, revision of diagnostic criteria, and new sensitive and specific tools for disease prognostication and management. Assessment of minimal residual disease (MRD) in response to therapy is one of these tools, as longer progression-free survival (PFS) is seen consistently among patients who have achieved MRD negativity. Current therapies lead to unprecedented frequency and depth of response, and next-generation flow and sequencing methods to measure MRD in bone marrow are in use and being developed with sensitivities in the range of 10-5 to 10-6 cells. These technologies may be combined with functional imaging to detect MRD outside of bone marrow. Moreover, immune profiling methods are being developed to better understand the immune environment in myeloma and response to immunomodulatory agents while methods for molecular profiling of myeloma cells and circulating DNA in blood are also emerging. With the continued development and standardization of these methodologies, MRD has high potential for use in gaining new drug approvals in myeloma. The FDA has outlined two pathways by which MRD could be qualified as a surrogate endpoint for clinical studies directed at obtaining accelerated approval for new myeloma drugs. Most importantly, better understanding of MRD should also contribute to better treatment monitoring. Potentially, MRD status could be used as a prognostic factor for making treatment decisions and for informing timing of therapeutic interventions. Clin Cancer Res; 23(15); 3980-93. ©2017 AACR.


Asunto(s)
ADN Tumoral Circulante/sangre , Mieloma Múltiple/sangre , Mieloma Múltiple/tratamiento farmacológico , Neoplasia Residual/sangre , Biomarcadores de Tumor/genética , Médula Ósea/efectos de los fármacos , Médula Ósea/patología , Supervivencia sin Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Mieloma Múltiple/complicaciones , Mieloma Múltiple/genética , Neoplasia Residual/inducido químicamente , Neoplasia Residual/genética , Selección de Paciente , Pronóstico
10.
Cancer Res ; 64(8): 2643-8, 2004 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15087371

RESUMEN

Malignant cells undergo anoikis as they encounter fluid shear stress during transit to a metastatic site. We postulated that intracellular nitric oxide (NO) contributes to this cell death by comparing the growth of human colorectal carcinoma cells in low fluid shear stress rotated three-dimensional (Rotated 3-D) cultures with growth in static three-dimensional (Static 3-D) cultures on nonadherent surfaces and with two-dimensional monolayer (Monolayer 2-D) cultures. NO, loss of microtubules, and apoptosis increased significantly in Rotated 3-D cultures within 10 min and persisted at 24 h, whereas inhibition of NO synthase decreased apoptosis and intracellular NO and prevented tubulin degradation. Thus, fluid shear stress and three-dimensional growth increases NO synthase and NO to cause tubulin breakdown and induce anoikis. Intracellular NO in malignant cells entering the circulation may be a novel target for metastasis by colorectal carcinoma.


Asunto(s)
Apoptosis/fisiología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Microtúbulos/metabolismo , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico/metabolismo , Antioxidantes/farmacología , Reactores Biológicos , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Humanos , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II , Especies Reactivas de Oxígeno/metabolismo , omega-N-Metilarginina/farmacología
11.
JAMA ; 294(21): 2703-11, 2005 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-16333005

RESUMEN

CONTEXT: A 1990 National Institutes of Health Consensus Conference recommended that patients with stage III colon cancer receive adjuvant chemotherapy because survival was improved in clinical trials in patients who received a 5-fluorouracil-based regimen. OBJECTIVE: To determine whether adjuvant chemotherapy is used in the community as a standard of practice that improves outcome and whether it failed to benefit any specific sets of patients. DESIGN, SETTING, AND PARTICIPANTS: Prospective data from 85 934 patients with stage III colon cancer from 560 hospital cancer registries were entered into the National Cancer Data Base between 1990 and 2002 and included standard clinical, pathological, and first course of treatment variables. MAIN OUTCOME MEASURES: Prevalence of adjuvant chemotherapy usage and 5-year survival in patients treated in US hospitals. RESULTS: Adjuvant chemotherapy use increased from 39% in 1991 to 64% in 2002 but was lower in black, female, and elderly patients. It improved 5-year survival from almost 8% in 1991 to more than 16% in 1997 compared with surgery alone. Adjuvant chemotherapy increases survival in elderly patients as much as it does in younger patients. However, the benefit of adjuvant chemotherapy in blacks and those with high-grade cancers is not as great. CONCLUSIONS: Adjuvant chemotherapy use has increased from 1990 to 2002 for patients with stage III colon cancer with an associated increase in 5-year survival of 16%. The benefit of adjuvant chemotherapy seems to be lower in black patients and high-grade cancers. Women have the same benefit but are less often treated. Elderly patients have the same benefit as younger patients but are less frequently treated. New options for adjuvant therapy in 2004-2005 may further improve the outcome of patients with stage III colon cancer.


Asunto(s)
Quimioterapia Adyuvante/estadística & datos numéricos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/etnología , Adyuvantes Inmunológicos/uso terapéutico , Factores de Edad , Anciano , Anciano de 80 o más Años , Antimetabolitos Antineoplásicos/uso terapéutico , Población Negra/estadística & datos numéricos , Neoplasias del Colon/mortalidad , Neoplasias del Colon/patología , Utilización de Medicamentos , Femenino , Fluorouracilo/uso terapéutico , Humanos , Levamisol/uso terapéutico , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Modelos de Riesgos Proporcionales , Factores Sexuales , Análisis de Supervivencia , Estados Unidos , Población Blanca/estadística & datos numéricos
12.
Clin Exp Metastasis ; 21(8): 709-17, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-16035616

RESUMEN

Most circulating tumor cells die within 24 h of entering the hepatic microvasculature because their arrest initiates an ischemia-reperfusion (I/R) injury that is cytotoxic. Human colorectal carcinomas (CRC) produce the glycoprotein Carcinoembryonic Antigen (CEA) that increases experimental liver metastasis in nude mice. Since CEA induces release of IL-6 and IL-10, we hypothesized that CEA inhibits the I/R injury through a Kupffer cell-mediated cytokine-dependent pathway. We assessed cytokine effects in CRC co-cultured with liver and in vivo. Human CRC prelabeled with fluorescent dyes were incubated with a reoxygenated suspension of ischemic nude mouse liver fragments in a bioreactor. CEA, rhIL-6 or rhIL-10 were either administered to the donor mice prior to hepatic ischemia or during co-culture. Liver donors were athymic nude or iNOS, IL-6 or IL-10 knock out mice. Ischemic-reoxygenated liver kills Clone A CRC through production of nitric oxide (NO) and superoxide anion. Treatment of liver donors with CEA prior to hepatic ischemia inhibited this in vitro cytotoxicity through an IL-10 and Kupffer cell dependent pathway that inhibited NF-kappaB activation, NO production and iNOS upregulation. IL-10 but not IL-6 enhanced CRC survival in nude mouse liver in vivo. Thus, CEA enhanced metastasis by inducing IL-10 to inhibit iNOS upregulation in host liver.


Asunto(s)
Antígeno Carcinoembrionario/farmacología , Neoplasias Colorrectales/patología , Interleucina-10/metabolismo , Neoplasias Hepáticas/secundario , Animales , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Neoplasias Colorrectales/metabolismo , Colorantes Fluorescentes , Humanos , Interleucina-6/genética , Interleucina-6/farmacología , Interleucina-6/fisiología , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , FN-kappa B/genética , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Estrés Oxidativo/efectos de los fármacos , Oxígeno/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Transducción de Señal , Superóxidos/metabolismo
13.
Clin Cancer Res ; 20(21): 5446-55, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25208882

RESUMEN

PURPOSE: High levels of BCL-2 family members in colorectal carcinoma cause resistance to treatment. Inhibition of NANOG or its paralog NANOGP8 reduces the proliferation, stemness, and tumorigenicity of colorectal carcinoma cells. Our hypothesis was that inhibition of NANOG/NANOGP8 enhances the cytotoxic effect of BH3 mimetics targeting BCL-2 family members in colorectal carcinoma cells through reducing expression of MCL-1, a prosurvival BCL-2 protein. EXPERIMENTAL DESIGN: Lentiviral vector (LV) shRNA to NANOG (shNG-1) or NANOGP8 (shNp8-1) transduced colorectal carcinoma cells that were also exposed to the BH3 mimetics ABT-737 or ABT-199 in vivo in colorectal carcinoma xenografts and in vitro where proliferation, protein and gene expression, and apoptosis were measured. RESULTS: Clone A and CX-1 were sensitive to ABT-737 and ABT-199 at IC50s of 2 to 9 µmol/L but LS174T was resistant with IC50s of 18 to 30 µmol/L. Resistance was associated with high MCL-1 expression in LS174T. LVshNG-1 or LVshNp8-1 decreased MCL-1 expression, increased apoptosis, and decreased replating efficiency in colorectal carcinoma cells treated with either ABT-737 or ABT-199 compared with the effects of either BH3 mimetic alone. Inhibition or overexpression of MCL-1 alone replicated the effects of LVshNG-1 or LVshNp8-1 in increasing or decreasing the apoptosis caused with the BH3 mimetic. The combination therapy inhibited the growth of LS174T xenografts in vivo compared with untreated controls or treatment with only LV shRNA or ABT-737. CONCLUSIONS: Inhibition of NANOGP8 or NANOG enhances the cytotoxicity of BH3 mimetics that target BCL-2 family members. Gene therapy targeting the NANOGs may increase the efficacy of BH3 mimetics in colorectal carcinoma.


Asunto(s)
Proteína Proapoptótica que Interacciona Mediante Dominios BH3/genética , Neoplasias Colorrectales/genética , Regulación hacia Abajo/genética , Proteínas de Homeodominio/antagonistas & inhibidores , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Compuestos de Bifenilo/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Línea Celular , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Regulación hacia Abajo/efectos de los fármacos , Células HEK293 , Células HT29 , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteína Homeótica Nanog , Nitrofenoles/farmacología , Piperazinas/farmacología , ARN Interferente Pequeño/genética , Sulfonamidas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Clin Cancer Res ; 20(6): 1428-44, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24634466

RESUMEN

This article defines and describes best practices for the academic and business community to generate evidence of clinical utility for cancer molecular diagnostic assays. Beyond analytical and clinical validation, successful demonstration of clinical utility involves developing sufficient evidence to demonstrate that a diagnostic test results in an improvement in patient outcomes. This discussion is complementary to theoretical frameworks described in previously published guidance and literature reports by the U.S. Food and Drug Administration, Centers for Disease Control and Prevention, Institute of Medicine, and Center for Medical Technology Policy, among others. These reports are comprehensive and specifically clarify appropriate clinical use, adoption, and payer reimbursement for assay manufacturers, as well as Clinical Laboratory Improvement Amendments-certified laboratories, including those that develop assays (laboratory developed tests). Practical criteria and steps for establishing clinical utility are crucial to subsequent decisions for reimbursement without which high-performing molecular diagnostics will have limited availability to patients with cancer and fail to translate scientific advances into high-quality and cost-effective cancer care. See all articles in this CCR Focus section, "The Precision Medicine Conundrum: Approaches to Companion Diagnostic Co-development."


Asunto(s)
Aprobación de Pruebas de Diagnóstico , Técnicas de Diagnóstico Molecular , Neoplasias/diagnóstico , Guías de Práctica Clínica como Asunto , Aprobación de Pruebas de Diagnóstico/normas , Aprobación de Pruebas de Diagnóstico/tendencias , Humanos , Técnicas de Diagnóstico Molecular/normas , Técnicas de Diagnóstico Molecular/tendencias , Terapia Molecular Dirigida/métodos , Neoplasias/terapia , Guías de Práctica Clínica como Asunto/normas , Estados Unidos
15.
Clin Cancer Res ; 18(6): 1531-9, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22422405

RESUMEN

The development of clinically useful molecular diagnostics requires validation of clinical assay performance and achievement of clinical qualification in clinical trials. As discussed elsewhere in this Focus section on molecular diagnostics, validation of assay performance must be rigorous, especially when the assay will be used to guide treatment decisions. Here we review some of the problems associated with assay development, especially for academic investigators. These include lack of expertise and resources for analytical validation, lack of experience in designing projects for a specific clinical use, lack of specimens from appropriate patient groups, and lack of access to Clinical Laboratory Improvement Amendments-certified laboratories. In addition, financial support for assay validation has lagged behind financial support for marker discovery or drug development, even though the molecular diagnostic may be considered necessary for the successful use of the companion therapeutic. The National Cancer Institute supports a large number of clinical trials and a significant effort in drug development. In order to address some of these barriers for predictive and prognostic assays that will be used in clinical trials to select patients for a particular treatment, stratify patients into molecularly defined subgroups, or choose between treatments for molecularly defined tumors, the National Cancer Institute has begun a pilot program designed to lessen barriers to the development of validated prognostic and predictive assays.


Asunto(s)
Biomarcadores de Tumor/análisis , Patología Molecular/normas , Investigación Biomédica , Ensayos Clínicos como Asunto , Humanos , National Institutes of Health (U.S.) , Neoplasias/diagnóstico , Proyectos Piloto , Pronóstico , Estados Unidos , Estudios de Validación como Asunto
16.
Clin Cancer Res ; 18(6): 1547-54, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22422407

RESUMEN

The U.S. Food and Drug Administration is now exerting its regulatory authority over the use of molecular diagnostics and related assays for medical decision making in clinical trials, by performing pre-Investigational Device Exemption reviews in all phases of clinical trials. In this review, we assess the analytical performance of the assay for the diagnostic, and consider how that performance affects the diagnostic and the patient and their risks and benefits from treatment. We also discuss the process involved in the first review of a new Children's Oncology Group phase III trial in acute myelogenous leukemia. The lessons learned and recommendations for how to prepare for and incorporate this new level of regulatory review into the protocol development process are presented.


Asunto(s)
Aprobación de Recursos , Regulación Gubernamental , Técnicas de Diagnóstico Molecular , United States Food and Drug Administration , Biomarcadores de Tumor/análisis , Ensayos Clínicos Fase III como Asunto , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/tratamiento farmacológico , Ensayos Clínicos Controlados Aleatorios como Asunto , Estados Unidos , Estudios de Validación como Asunto
17.
PLoS One ; 7(2): e31058, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22347427

RESUMEN

Exposure to oxygen-rich environments can lead to oxidative damage, increased body iron stores, and changes in status of some vitamins, including folate. Assessing the type of oxidative damage in these environments and determining its relationships with changes in folate status are important for defining nutrient requirements and designing countermeasures to mitigate these effects. Responses of humans to oxidative stressors were examined in participants undergoing a saturation dive in an environment with increased partial pressure of oxygen, a NASA Extreme Environment Mission Operations mission. Six participants completed a 13-d saturation dive in a habitat 19 m below the ocean surface near Key Largo, FL. Fasting blood samples were collected before, twice during, and twice after the dive and analyzed for biochemical markers of iron status, oxidative damage, and vitamin status. Body iron stores and ferritin increased during the dive (P<0.001), with a concomitant decrease in RBC folate (P<0.001) and superoxide dismutase activity (P<0.001). Folate status was correlated with serum ferritin (Pearson r = -0.34, P<0.05). Peripheral blood mononuclear cell poly(ADP-ribose) increased during the dive and the increase was significant by the end of the dive (P<0.001); γ-H2AX did not change during the mission. Together, the data provide evidence that when body iron stores were elevated in a hyperoxic environment, a DNA damage repair response occurred in peripheral blood mononuclear cells, but double-stranded DNA damage did not. In addition, folate status decreases quickly in this environment, and this study provides evidence that folate requirements may be greater when body iron stores and DNA damage repair responses are elevated.


Asunto(s)
Daño del ADN , Reparación del ADN , Buceo/efectos adversos , Ácido Fólico/sangre , Biomarcadores/sangre , Análisis Químico de la Sangre , Humanos , Hiperoxia , Hierro/análisis , Oxígeno , Estados Unidos , United States National Aeronautics and Space Administration
18.
Clin Cancer Res ; 18(6): 1515-23, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22422403

RESUMEN

Molecular diagnostics are becoming increasingly important in clinical research to stratify or identify molecularly profiled patient cohorts for targeted therapies, to modify the dose of a therapeutic, and to assess early response to therapy or monitor patients. Molecular diagnostics can also be used to identify the pharmacogenetic risk of adverse drug reactions. The articles in this CCR Focus section on molecular diagnosis describe the development and use of markers to guide medical decisions regarding cancer patients. They define sources of preanalytic variability that need to be minimized, as well as the regulatory and financial challenges involved in developing diagnostics and integrating them into clinical practice. They also outline a National Cancer Institute program to assist diagnostic development. Molecular diagnostic clinical tests require rigor in their development and clinical validation, with sensitivity, specificity, and validity comparable to those required for the development of therapeutics. These diagnostics must be offered at a realistic cost that reflects both their clinical value and the costs associated with their development. When genome-sequencing technologies move into the clinic, they must be integrated with and traceable to current technology because they may identify more efficient and accurate approaches to drug development. In addition, regulators may define progressive drug approval for companion diagnostics that requires further evidence regarding efficacy and safety before full approval can be achieved. One way to accomplish this is to emphasize phase IV postmarketing, hypothesis-driven clinical trials with biological characterization that would permit an accurate definition of the association of low-prevalence gene alterations with toxicity or response in large cohorts.


Asunto(s)
Biomarcadores de Tumor , Diseño de Fármacos , Neoplasias/diagnóstico , Patología Molecular/normas , Bancos de Muestras Biológicas , Ensayos Clínicos como Asunto , Bases de Datos como Asunto , Humanos , Técnicas de Diagnóstico Molecular , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Estudios de Validación como Asunto
19.
J Clin Oncol ; 30(19): 2327-33, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22585691

RESUMEN

PURPOSE: Surgical resection of gastric cancer has produced suboptimal survival despite multiple randomized trials that used postoperative chemotherapy or more aggressive surgical procedures. We performed a randomized phase III trial of postoperative radiochemotherapy in those at moderate risk of locoregional failure (LRF) following surgery. We originally reported results with 4-year median follow-up. This update, with a more than 10-year median follow-up, presents data on failure patterns and second malignancies and explores selected subset analyses. PATIENTS AND METHODS: In all, 559 patients with primaries ≥ T3 and/or node-positive gastric cancer were randomly assigned to observation versus radiochemotherapy after R0 resection. Fluorouracil and leucovorin were administered before, during, and after radiotherapy. Radiotherapy was given to all LRF sites to a dose of 45 Gy. RESULTS: Overall survival (OS) and relapse-free survival (RFS) data demonstrate continued strong benefit from postoperative radiochemotherapy. The hazard ratio (HR) for OS is 1.32 (95% CI, 1.10 to 1.60; P = .0046). The HR for RFS is 1.51 (95% CI, 1.25 to 1.83; P < .001). Adjuvant radiochemotherapy produced substantial reduction in both overall relapse and locoregional relapse. Second malignancies were observed in 21 patients with radiotherapy versus eight with observation (P = .21). Subset analyses show robust treatment benefit in most subsets, with the exception of patients with diffuse histology who exhibited minimal nonsignificant treatment effect. CONCLUSION: Intergroup 0116 (INT-0116) demonstrates strong persistent benefit from adjuvant radiochemotherapy. Toxicities, including second malignancies, appear acceptable, given the magnitude of RFS and OS improvement. LRF reduction may account for the majority of overall relapse reduction. Adjuvant radiochemotherapy remains a rational standard therapy for curatively resected gastric cancer with primaries T3 or greater and/or positive nodes.


Asunto(s)
Adenocarcinoma/terapia , Gastrectomía/métodos , Neoplasias Gástricas/cirugía , Neoplasias Gástricas/terapia , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/radioterapia , Adenocarcinoma/cirugía , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Quimioradioterapia Adyuvante , Femenino , Fluorouracilo/administración & dosificación , Humanos , Leucovorina/administración & dosificación , Masculino , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/radioterapia , Análisis de Supervivencia , Resultado del Tratamiento
20.
Clin Cancer Res ; 16(6): 1745-55, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20215558

RESUMEN

The National Cancer Institute (NCI) Investigational Drug Steering Committee (IDSC) charged the Biomarker Task Force to develop recommendations to improve the decisions about incorporation of biomarker studies in early investigational drug trials. The Task Force members reviewed biomarker trials, the peer-reviewed literature, NCI and U.S. Food and Drug Administration (FDA) guidance documents, and conducted a survey of investigators to determine practices and challenges to executing biomarker studies in clinical trials of new drugs in early development. This document provides standard definitions and categories of biomarkers, and lists recommendations to sponsors and investigators for biomarker incorporation into such trials. Our recommendations for sponsors focus on the identification and prioritization of biomarkers and assays, the coordination of activities for the development and use of assays, and for operational activities. We also provide recommendations for investigators developing clinical trials with biomarker studies for scientific rationale, assay criteria, trial design, and analysis. The incorporation of biomarker studies into early drug trials is complex. Thus the decision to proceed with studies of biomarkers should be based on balancing the strength of science, assay robustness, feasibility, and resources with the burden of proper sample collection on the patient and potential impact of the results on drug development. The Task Force provides these guidelines in the hopes that improvements in biomarker studies will enhance the efficiency of investigational drug development.


Asunto(s)
Biomarcadores de Tumor/análisis , Ensayos Clínicos como Asunto/métodos , Ensayos Clínicos como Asunto/normas , Drogas en Investigación/normas , Neoplasias/tratamiento farmacológico , Comité Farmacéutico y Terapéutico/organización & administración , Proyectos de Investigación , Determinación de Punto Final , Humanos , National Cancer Institute (U.S.) , Neoplasias/química , Seguridad , Estados Unidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA