Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Cells ; 12(18)2023 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-37759464

RESUMEN

The lack of optimal models to evaluate novel agents is delaying the development of effective immunotherapies against human breast cancer (BC). In this prospective open label study, we applied neoadjuvant intratumoral immunotherapy with empty cowpea mosaic virus-like particles (eCPMV) to 11 companion dogs diagnosed with canine mammary cancer (CMC), a spontaneous tumor resembling human BC. We found that two neoadjuvant intratumoral eCPMV injections resulted in tumor reduction in injected tumors in all patients and in noninjected tumors located in the ipsilateral and contralateral mammary chains of injected dogs. Tumor reduction was independent of clinical stage, tumor size, histopathologic grade, and tumor molecular subtype. RNA-seq-based analysis of injected tumors indicated a decrease in DNA replication activity and an increase in activated dendritic cell infiltration in the tumor microenvironment. Immunohistochemistry analysis demonstrated significant intratumoral increases in neutrophils, T and B lymphocytes, and plasma cells. eCPMV intratumoral immunotherapy demonstrated antitumor efficacy without any adverse effects. This novel immunotherapy has the potential for improving outcomes for human BC patients.


Asunto(s)
Neoplasias de la Mama , Comovirus , Humanos , Animales , Perros , Femenino , Terapia Neoadyuvante , Estudios Prospectivos , Neoplasias de la Mama/terapia , Inmunoterapia , Microambiente Tumoral
2.
Cells ; 11(18)2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-36139425

RESUMEN

Inflammatory breast cancer (IBC) is highly metastatic at the onset of the disease with no IBC-specific treatments, resulting in dismal patient survival. IBC treatment is a clear unmet clinical need. This commentary highlights findings from a recent seminal approach in which pembrolizumab, a checkpoint inhibitor against programmed cell death protein 1 (PD-1), was provided to a triple-negative IBC patient as a neoadjuvant immune therapy combined with anthracycline-taxane-based chemotherapy. We highlight the findings of the case report and offer a perspective on taking a proactive approach to deploy approved immune checkpoint inhibitors. On the basis of our recently published research study, we propose in situ vaccination with direct injection of immunostimulatory agents into the tumor as an option to improve outcomes safely, effectively, and economically for IBC patients.


Asunto(s)
Neoplasias Inflamatorias de la Mama , Antraciclinas , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia , Neoplasias Inflamatorias de la Mama/patología , Neoplasias Inflamatorias de la Mama/terapia , Receptor de Muerte Celular Programada 1 , Taxoides
3.
J Immunother Cancer ; 10(3)2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35277459

RESUMEN

BACKGROUND: Inflammatory mammary cancer (IMC), the counterpart of human inflammatory breast cancer (IBC), is the deadliest form of canine mammary tumors. IMC patients lack specific therapy and have poor outcomes. This proof-of-principle preclinical study evaluated the efficacy, safety, and effect on survival of neoadjuvant intratumoral (in situ) empty cowpea mosaic virus (eCPMV) immunotherapy in companion dogs diagnosed with IMC. METHODS: Ten IMC-bearing dogs were enrolled in the study. Five dogs received medical therapy, and five received weekly neoadjuvant in situ eCPMV immunotherapy (0.2-0.4 mg per injection) and medical therapy after the second eCPMV injection. Efficacy was evaluated by reduction of tumor growth; safety by hematological and biochemistry changes in blood and plasma; and patient outcome by survival analysis. eCPMV-induced immune changes in blood cells were analyzed by flow cytometry; changes in the tumor microenvironment were evaluated by CD3 (T lymphocytes), CD20 (B lymphocytes), FoxP3 (Treg lymphocytes), myeloperoxidase (MPO; neutrophils), Ki-67 (proliferation index, PI; tumor cell proliferation), and Cleaved Caspase-3 (CC-3; apoptosis) immunohistochemistry. RESULTS: Two neoadjuvant in situ eCPMV injections resulted in tumor shrinkage in all patients by day 14 without systemic adverse events. Although surgery for IMC is generally not an option, reduction in tumor size allowed surgery in two IMC patients. In peripheral blood, in situ eCPMV immunotherapy was associated with a significant decrease of Treg+/CD8+ ratio and changes in CD8+Granzyme B+ T cells, which behave as a lagging predictive biomarker. In the TME, higher neutrophilic infiltration and MPO expression, lower tumor Ki-67 PI, increase in CD3+ lymphocytes, decrease in FoxP3+/CD3+ ratio (p<0.04 for all comparisons), and no changes in CC-3+ immunostainings were observed in post-treatment tumor tissues when compared with pretreatment tumor samples. eCPMV-treated IMC patients had a statistically significant (p=0.033) improved overall survival than patients treated with medical therapy. CONCLUSIONS: Neoadjuvant in situ eCPMV immunotherapy demonstrated anti-tumor efficacy and improved survival in IMC patients without systemic adverse effects. eCPMV-induced changes in immune cells point to neutrophils as a driver of immune response. Neoadjuvant in situ eCPMV immunotherapy could be a groundbreaking immunotherapy for canine IMC and a potential future immunotherapy for human IBC patients.


Asunto(s)
Comovirus , Neoplasias Inflamatorias de la Mama , Neoplasias , Animales , Perros , Factores de Transcripción Forkhead , Humanos , Antígeno Ki-67 , Terapia Neoadyuvante , Microambiente Tumoral , Vacunación
4.
Breast Cancer Res Treat ; 190(3): 477-489, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34542773

RESUMEN

PURPOSE: This study evaluated epidemiologic and immune factors associated with pathologic complete response (pCR), breast cancer-specific survival (BCSS) and disease-free survival (DFS) outcomes in inflammatory (IBC) and locally advanced breast cancer (LABC) patients. METHODS: Tumor-infiltrating lymphocytes (TILs) and CD20+ B-cell frequencies (CD20+), and PD-L1 expression on tumor (PD-L1+carcinoma cells) and immune (PD-L1+TILs) cells were analyzed by immunohistochemistry along with clinicopathologic factors as modifiers of pCR and outcomes in 221 IBC and 162 LABC patients. Analysis included Kaplan-Meier curves and Cox proportional hazard models. RESULTS: IBC and LABC display similar levels of TILs, CD20+, and combined CD20+ and PD-L1+TILs (CD20+PD-L1+TILs), while LABC contained more PD-L1+TILs and PD-L1+ carcinoma cells. Absence of lymphovascular involvement, high TILs, PD-L1+ carcinoma cells, and combined CD20+ and PD-L1+ carcinoma cells correlated with pCR in IBC and LABC patients. High PD-L1+TILs correlated with pCR only in LABC; less lymph node involvement at diagnosis, CD20+ and CD20+PD-L1+TILs correlated with pCR only in IBC (P < 0.04, all comparisons). Achievement of pCR in IBC and LABC patients correlated with BCSS and DFS (P < 0.02). In multivariate analyses, pCR remained an independent prognostic factor of improved DFS in IBC and LABC patients, but of BCSS in only LABC. CD20+PD-L1+TILs remained an independent prognostic factor of improved DFS and BCSS only in IBC. CONCLUSION: CD20+PD-L1+TILs are an independent prognostic biomarker of improved outcomes in IBC, but not LABC. Selecting IBC patients by CD20 and PD-L1 status could stratify patients and potentially identify those in whom activating CD20 agents and anti-PD-1/PD-L1 therapy could be explored.


Asunto(s)
Neoplasias de la Mama , Neoplasias Inflamatorias de la Mama , Antígenos CD20 , Linfocitos B , Antígeno B7-H1/genética , Biomarcadores de Tumor , Neoplasias de la Mama/tratamiento farmacológico , Femenino , Humanos , Linfocitos Infiltrantes de Tumor , Pronóstico
5.
Nat Commun ; 12(1): 2804, 2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33990571

RESUMEN

Chemotherapy remains the standard of care for most cancers worldwide, however development of chemoresistance due to the presence of the drug-effluxing ATP binding cassette (ABC) transporters remains a significant problem. The development of safe and effective means to overcome chemoresistance is critical for achieving durable remissions in many cancer patients. We have investigated the energetic demands of ABC transporters in the context of the metabolic adaptations of chemoresistant cancer cells. Here we show that ABC transporters use mitochondrial-derived ATP as a source of energy to efflux drugs out of cancer cells. We further demonstrate that the loss of methylation-controlled J protein (MCJ) (also named DnaJC15), an endogenous negative regulator of mitochondrial respiration, in chemoresistant cancer cells boosts their ability to produce ATP from mitochondria and fuel ABC transporters. We have developed MCJ mimetics that can attenuate mitochondrial respiration and safely overcome chemoresistance in vitro and in vivo. Administration of MCJ mimetics in combination with standard chemotherapeutic drugs could therefore become an alternative strategy for treatment of multiple cancers.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Adenosina Trifosfato/metabolismo , Resistencia a Antineoplásicos/fisiología , Mitocondrias/metabolismo , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Línea Celular Tumoral , Respiración de la Célula/efectos de los fármacos , Respiración de la Célula/fisiología , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Resistencia a Múltiples Medicamentos/fisiología , Femenino , Proteínas del Choque Térmico HSP40/deficiencia , Proteínas del Choque Térmico HSP40/metabolismo , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mitocondrias/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Consumo de Oxígeno/efectos de los fármacos
6.
Semin Oncol ; 44(4): 288-300, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-29526258

RESUMEN

Inflammatory breast cancer (IBC) in humans is the most aggressive form of mammary gland cancer and shares clinical, pathologic, and molecular patterns of disease with canine inflammatory mammary carcinoma (CIMC). Despite the use of multimodal therapeutic approaches, including targeted therapies, the prognosis for IBC/CIMC remains poor. The aim of this review is to critically analyze IBC and CIMC in terms of biology and clinical features. While rodent cancer models have formed the basis of our understanding of cancer biology, the translation of this knowledge into improved outcomes has been limited. However, it is possible that a comparative "one health" approach to research, using a natural canine model of the disease, may help advance our knowledge on the biology of the disease. This will translate into better clinical outcomes for both species. We propose that CIMC has the potential to be a useful model for developing and testing novel therapies for IBC. Further, this strategy could significantly improve and accelerate the design and establishment of new clinical trials to identify novel and improved therapies for this devastating disease in a more predictable way.


Asunto(s)
Enfermedades de los Perros/terapia , Neoplasias Inflamatorias de la Mama/etiología , Neoplasias Inflamatorias de la Mama/veterinaria , Animales , Enfermedades de los Perros/etiología , Perros , Familia de Proteínas EGF/genética , Familia de Proteínas EGF/metabolismo , Femenino , Humanos , Neoplasias Inflamatorias de la Mama/patología , Neoplasias Inflamatorias de la Mama/terapia , Pronóstico , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo
7.
Immunology ; 150(1): 25-34, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27564847

RESUMEN

The role of the immune system in shaping cancer development and patient prognosis has recently become an area of intense focus in industry and academia. Harnessing the adaptive arm of the immune system for tumour eradication has shown great promise in a variety of tumour types. Differences between tissues, however, necessitate a greater understanding of the adaptive immunity programmes that are active within each tumour type. In breast cancer, adaptive immune programmes play diverse roles depending on the cellular infiltration found in each tumour. Cytotoxic T lymphocytes and T helper type 1 cells can induce tumour eradication, whereas regulatory T cells and T helper type 2 cells are known to be involved in tumour-promoting immunosuppressive responses. Complicating these matters, heterogeneous expression of hormone receptors and growth factors in different tumours leads to disparate, patient-specific adaptive immune responses. Despite this non-conformity in adaptive immune behaviours, encouraging basic and clinical results have been observed that suggest a role for immunotherapeutic approaches in breast cancer. Here, we review the literature pertaining to the adaptive immune response in breast cancer, summarize the primary findings relating to the breast tumour's biology, and discuss potential clinical immunotherapies.


Asunto(s)
Inmunidad Adaptativa , Neoplasias de la Mama/inmunología , Inmunoterapia Adoptiva/métodos , Linfocitos T/inmunología , Animales , Femenino , Humanos
8.
Genes Chromosomes Cancer ; 55(2): 177-89, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26542757

RESUMEN

Multiple chromosomal regions are affected by deletions in cervical cancer (CC) genomes, but their consequence and target gene involvement remains unknown. Our single nucleotide polymorphism (SNP) array identified 8p copy number losses localized to an 8.4 Mb minimal deleted region (MDR) in 36% of CC. The 8p MDR was associated with tumor size, treatment outcome, and with multiple HPV infections. Genetic, epigenetic, and expression analyses of candidate genes at MDR identified promoter hypermethylation and/or inactivation of decoy receptors TNFRSF10C and TNFRSF10D in the majority of CC patients. TNFRSF10C methylation was also detected in precancerous lesions suggesting that this change is an early event in cervical tumorigenesis. We further demonstrate here that CC cell lines exhibiting downregulated expression of TNFRSF10C and/or TNFRSF10D effectively respond to TRAIL-induced apoptosis and this affect was synergistic in combination with DNA damaging chemotherapeutic drugs. We show that the CC cell lines harboring epigenetic inactivation of TRAIL decoy receptors effectively activate downstream caspases suggesting a critical role of inactivation of these genes in efficient execution of extrinsic apoptotic pathway and therapy response. Therefore, these findings shed new light on the role of genetic/epigenetic defects in TRAIL decoy receptor genes in the pathogenesis of CC and provide an opportunity to explore strategies to test decoy receptor gene inactivation as a biomarker of response to Apo2L/TRAIL-combination therapy.


Asunto(s)
Cisplatino/farmacología , Metilación de ADN , Miembro 10c de Receptores del Factor de Necrosis Tumoral/genética , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Receptores Señuelo del Factor de Necrosis Tumoral/genética , Neoplasias del Cuello Uterino/tratamiento farmacológico , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica , Secuencia de Bases , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cromosomas Humanos Par 8/genética , Cisplatino/uso terapéutico , Epigénesis Genética , Femenino , Proteínas Ligadas a GPI/genética , Células HeLa , Humanos , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Eliminación de Secuencia , Neoplasias del Cuello Uterino/genética
10.
Cancer Metastasis Rev ; 33(1): 183-215, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24338006

RESUMEN

Oxygen is the basic molecule which supports life and it truly is "god's gift to life." Despite its immense importance, research on "oxygen biology" has never received the light of the day and has been limited to physiological and biochemical studies. It seems that in modern day biology, oxygen research is summarized in one word "hypoxia." Scientists have focused on hypoxia-induced transcriptomics and molecular-cellular alterations exclusively in disease models. Interestingly, the potential of oxygen to control the basic principles of biology like homeostatic maintenance, transcription, replication, and protein folding among many others, at the molecular level, has been completely ignored. Here, we present a perspective on the crucial role played by oxygen in regulation of basic biological phenomena. Our conclusion highlights the importance of establishing novel research areas like oxygen biology, as there is great potential in this field for basic science discoveries and clinical benefits to the society.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Neoplasias/metabolismo , Oxígeno/metabolismo , Hipoxia de la Célula , Línea Celular Tumoral , Progresión de la Enfermedad , Humanos , Hipoxia , Modelos Genéticos , Metástasis de la Neoplasia , Neoplasias/patología
11.
Oral Oncol ; 49(5): 461-7, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23384718

RESUMEN

OBJECTIVES: The epidermal growth factor receptor (EGFR) is a validated target in head and neck squamous cell carcinoma (HNSCC). In recurrent and/or metastatic (R/M) HNSCC, resistance to anti-EGFR therapy inevitably occurs. Downstream activation of the PI3K/Akt/mTOR pathway is an established resistance mechanism. Concurrent mTOR blockade may improve efficacy of anti-EGFR therapy. MATERIALS AND METHODS: Erlotinib 150 mg daily and temsirolimus 15 mg weekly were administered to patients with platinum-refractory R/M HNSCC and ECOG performance status 0-2. The primary endpoint was progression-free survival (PFS). Correlative studies determined PIK3CA and HRAS mutation status; p16, EGFR, pS6K, pAkt and PTEN expression; and pre- and post-treatment plasma levels of 20 immunomodulatory cytokines. RESULTS: Twelve patients enrolled; six withdrew within 6 weeks due to toxicity or death, prompting early closure of the trial. Grade ≥ 3 toxicities included fatigue, diarrhea, gastrostomy tube infection, peritonitis, pneumonia, dyspnea, and HN edema. Median PFS was 1.9 months. Median overall survival was 4.0 months. Six/12 tumors were p16(+), 9/11 lacked measurable PTEN expression, and 1/12 harbored a PIK3CA mutation. On exploratory analysis, high baseline plasma VEGF and interferon-gamma levels marginally associated with tumor progression. CONCLUSIONS: The combination of erlotinib and temsirolimus was poorly tolerated. Low prevalence of PTEN expression and 8% incidence of PIK3CA mutations indicate biological relevance of this pathway in R/M disease. Investigation of more tolerable combinations of EGFR and PI3K/Akt/mTOR pathway inhibitors in selected HNSCC patients is warranted.


Asunto(s)
Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Receptores ErbB/antagonistas & inhibidores , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/administración & dosificación , Quinazolinas/administración & dosificación , Sirolimus/análogos & derivados , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carcinoma de Células Escamosas/secundario , Fosfatidilinositol 3-Quinasa Clase I , Inhibidor p16 de la Quinasa Dependiente de Ciclina/análisis , Citocinas/análisis , Resistencia a Antineoplásicos , Clorhidrato de Erlotinib , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación/genética , Proteína Oncogénica v-akt/análisis , Fosfohidrolasa PTEN/análisis , Fosfatidilinositol 3-Quinasas/análisis , Fosfatidilinositol 3-Quinasas/genética , Platino (Metal) , Proteínas Proto-Oncogénicas p21(ras)/análisis , Proteínas Proto-Oncogénicas p21(ras)/genética , Quinazolinas/efectos adversos , Proteínas Quinasas S6 Ribosómicas/análisis , Sirolimus/administración & dosificación , Sirolimus/efectos adversos , Tasa de Supervivencia , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Proteínas Supresoras de Tumor/análisis
12.
BMC Cancer ; 12: 449, 2012 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-23033932

RESUMEN

BACKGROUND: In non-small cell lung cancer (NSCLC), interstitial hypertension is a barrier to chemotherapy delivery, and is mediated by platelet derived growth factor receptor (PDGFR). Antagonizing PDGFR with imatinib may improve intra-tumoral delivery of paclitaxel, increasing response rate (RR). METHODS: This single-stage, open-label phase II study evaluated pulse dose imatinib and weekly paclitaxel in elderly patients with advanced NSCLC. Eligible patients were aged ≥ 70 with untreated, stage IIIB-IV NSCLC and ECOG performance status 0-2. Primary endpoint was RR. Secondary endpoints included median progression free and overall survival (PFS, OS) and correlatives of PDGFR pathway activation. Baseline Charlson Comorbidity Index (CCI) and Vulnerable Elder Survey-13 (VES-13) were correlated with outcomes. RESULTS: Thirty-four patients with median age 75 enrolled. Eleven of 29 (38%) were frail by VES-13 score. Overall RR was 11/34 (32%; 95% CI 17%-51%), meeting the primary endpoint. Median PFS and OS were 3.6 and 7.3 months, respectively. High tumoral PDGF-B expression predicted inferior PFS. Frail patients by VES-13 had significantly worse median PFS (3.2 vs. 4.5 months; p=0.02) and OS (4.8 vs. 12 months; p=0.02) than non-frail. CONCLUSIONS: The combination of imatinib and paclitaxel had encouraging activity as measured by the primary endpoint of RR. However, PFS and OS were typical for elderly patients treated with single agent chemotherapy and the regimen is not recommended for further study. Adjunct imatinib did not overcome the established association of tumoral PDGF-B expression with inferior PFS. VES-13 was a powerful predictor of poor survival outcomes. Frailty should be further studied as a predictor of non-benefit from chemotherapy. TRIAL REGISTRATION: ClinicalTrials.gov NCT01011075.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Pulmón/efectos de los fármacos , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Benzamidas/administración & dosificación , Benzamidas/efectos adversos , Carcinoma de Pulmón de Células no Pequeñas/patología , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Fatiga/inducido químicamente , Femenino , Anciano Frágil , Humanos , Mesilato de Imatinib , Pulmón/metabolismo , Pulmón/patología , Neoplasias Pulmonares/patología , Masculino , Estadificación de Neoplasias , Neutropenia/inducido químicamente , Paclitaxel/administración & dosificación , Paclitaxel/efectos adversos , Piperazinas/administración & dosificación , Piperazinas/efectos adversos , Proteínas Proto-Oncogénicas c-sis/metabolismo , Pirimidinas/administración & dosificación , Pirimidinas/efectos adversos , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Inducción de Remisión , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia , Resultado del Tratamiento
13.
BMC Cancer ; 12: 298, 2012 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-22813402

RESUMEN

BACKGROUND: Inflammatory breast cancer (IBC) is a highly angiogenic disease; thus, antiangiogenic therapy should result in a clinical response. However, clinical trials have demonstrated only modest responses, and the reasons for these outcomes remain unknown. Therefore, the purpose of this retrospective study was to determine the prognostic value of protein levels of vascular endothelial growth factor (VEGF-A), one of the main targets of antiangiogenic therapy, and its receptors (VEGF-R1 and -R2) in IBC tumor specimens. PATIENTS AND METHODS: Specimens from IBC and normal breast tissues were obtained from Algerian patients. Tumor epithelial and stromal staining of VEGF-A, VEGF-R1, and VEGF-R2 was evaluated by immunohistochemical analysis in tumors and normal breast tissues; this expression was correlated with clinicopathological variables and breast cancer-specific survival (BCSS) and disease-free survival (DFS) duration. RESULTS: From a set of 117 IBC samples, we evaluated 103 ductal IBC tissues and 25 normal specimens. Significantly lower epithelial VEGF-A immunostaining was found in IBC tumor cells than in normal breast tissues (P <0.01), cytoplasmic VEGF-R1 and nuclear VEGF-R2 levels were slightly higher, and cytoplasmic VEGF-R2 levels were significantly higher (P = 0.04). Sixty-two percent of IBC tumors had high stromal VEGF-A expression. In univariate analysis, stromal VEGF-A levels predicted BCSS and DFS in IBC patients with estrogen receptor-positive (P <0.01 for both), progesterone receptor-positive (P = 0.04 and P = 0.03), HER2+ (P = 0.04 and P = 0.03), and lymph node involvement (P <0.01 for both). Strikingly, in a multivariate analysis, tumor stromal VEGF-A was identified as an independent predictor of poor BCSS (hazard ratio [HR]: 5.0; 95% CI: 2.0-12.3; P <0.01) and DFS (HR: 4.2; 95% CI: 1.7-10.3; P <0.01). CONCLUSIONS: To our knowledge, this is the first study to demonstrate that tumor stromal VEGF-A expression is a valuable prognostic indicator of BCSS and DFS at diagnosis and can therefore be used to stratify IBC patients into low-risk and high-risk groups for death and relapses. High levels of tumor stromal VEGF-A may be useful for identifying IBC patients who will benefit from anti-angiogenic treatment.


Asunto(s)
Neoplasias Inflamatorias de la Mama/diagnóstico , Neoplasias Inflamatorias de la Mama/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Femenino , Humanos , Neoplasias Inflamatorias de la Mama/mortalidad , Estimación de Kaplan-Meier , Estadificación de Neoplasias , Pronóstico , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Estudios Retrospectivos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
14.
PLoS One ; 7(5): e36525, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22567163

RESUMEN

BACKGROUND: The Death Receptor 6 (DR6) protein is elevated in the serum of ovarian cancer patients. We tested DR6 serum protein levels as a diagnostic/predictive biomarker in several epithelial tumors and sarcomas. METHODS: DR6 gene expression profiles were screened in publically available arrays of solid tumors. A quantitative immunofluorescent western blot analysis was developed to test the serum of healthy controls and patients with sarcoma, uterine carcinosarcoma, bladder, liver, and pancreatic carcinomas. Change in DR6 serum levels was used to assay the ability of DR6 to predict the response to therapy of sarcoma patients. RESULTS: DR6 mRNA is highly expressed in all tumor types assayed. Western blot analysis of serum DR6 protein demonstrated high reproducibility (r = 0.97). Compared to healthy donor controls, DR6 serum levels were not elevated in patients with uterine carcinosarcoma, bladder, liver, or pancreatic cancers. Serum DR6 protein levels from adult sarcoma patients were significantly elevated (p<0.001). This was most evident for patients with synovial sarcoma. Change in serum DR6 levels during therapy correlated with clinical benefit from therapy (sensitivity 75%, and positive predictive value 87%). CONCLUSION: DR6 may be a clinically useful diagnostic and predictive serum biomarker for some adult sarcoma subtypes. IMPACT: Diagnosis of sarcoma can be difficult and can lead to improper management of these cancers. DR6 serum protein may be a tool to aid in the diagnosis of some sarcomatous tumors to improve treatment planning. For patients with advanced disease, rising DR6 levels predict non-response to therapy and may expedite therapeutic decision making and reduce reliance on radiologic imaging.


Asunto(s)
Biomarcadores de Tumor/sangre , Receptores del Factor de Necrosis Tumoral/sangre , Sarcoma/sangre , Western Blotting , Femenino , Humanos , Neoplasias Hepáticas/sangre , Neoplasias Ováricas/sangre , Neoplasias Pancreáticas/sangre , Valor Predictivo de las Pruebas , Neoplasias Uterinas/sangre
15.
Breast Cancer Res Treat ; 131(2): 437-44, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21360074

RESUMEN

Inflammatory breast cancer (IBC) shows a high incidence in Tunisia and Egypt but epidemiological and molecular characteristics have not been described in Algeria. We compared 117 IBC and 59 non-IBC locally advanced breast cancers (LABC), for estrogen and progesterone receptors, HER2, and EGFR protein expression by immunohistochemistry, and HER2 gene amplification by chromogenic in situ hybridization. Demographic, clinico-pathological, and molecular variables were compared with chi-square and Fisher's exact tests to test for significance (P < 0.05, two-tailed). Overall survival (OS) and disease-free survival (DFS) were plotted using Kaplan-Meier curves and compared using the log-rank test. Tumor emboli were detected in 77% of IBC. Palpable masses were found in all LABC but only in 32% of IBC (P < 0.001). Recurrences were higher in LABC than in IBC (48 vs. 35%; P = 0.14) but OS was worse in IBC (68 vs. 71%; P = 0.06). There were no significant differences between IBC and LABC by demographics or by clinico-pathological parameters. The majority of IBC and LABC tumors were luminal A (62 and 64%), followed by basal (~18%, each), triple negative (~18%, each), and HER2+ (~10%, each) subtypes. In multivariate analyses, grade was associated with worse OS (P = 0.04), and DFS (P < 0.001) in IBC; chemo- and radio-therapy were associated with improved OS and DFS, respectively (P < 0.05 for each) in LABC. In conclusion, IBC in Algeria shows similar characteristics to IBC described for Egypt and Tunisia with subtle molecular differences. Current therapeutic treatments were not very effective in this population and new approaches are much needed.


Asunto(s)
Neoplasias Inflamatorias de la Mama/epidemiología , Neoplasias Inflamatorias de la Mama/genética , Adulto , Anciano , Anciano de 80 o más Años , Argelia/epidemiología , Biomarcadores de Tumor/genética , Femenino , Humanos , Neoplasias Inflamatorias de la Mama/terapia , Persona de Mediana Edad , Estadificación de Neoplasias , Recurrencia , Resultado del Tratamiento
16.
Int J Surg Pathol ; 19(5): 588-96, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21632639

RESUMEN

INTRODUCTION: Our study attempts to characterize mesothelin and GPR30 / estrogen receptor (ER) staining in pancreatic pathology. MATERIALS AND METHODS: Immunohistochemical staining for mesothelin, GPR30, and ER was performed on a variety of pancreatic lesions. RESULTS: 24 of 42 (57%) adenocarcinomas stained for mesothelin, while 0 of 16 non-carcinomas (0%) stained (p = 0.0000784). 35 of 39 (90%) adenocarcinomas stained for GPR30, while only 4 of 15 (27%) non-carcinomas stained (p = 0.0000036). Apart from stromal staining in one case of mucinous cystic neoplasm, no cases stained for ER. 27 of 37 (73%) adenocarcinoma fine needle aspirates were positive for mesothelin. DISCUSSION: GPR30 is more sensitive, but less specific than mesothelin for pancreatic adenocarcinoma. Mesothelin is detected in most adenocarcinoma fine needle aspirates. ER is rarely detected in pancreatic lesions.


Asunto(s)
Biomarcadores de Tumor/análisis , Proteínas Ligadas a GPI/análisis , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores de Estrógenos/análisis , Receptores Acoplados a Proteínas G/análisis , Biopsia con Aguja , Proteínas Ligadas a GPI/biosíntesis , Humanos , Inmunohistoquímica , Mesotelina , Clasificación del Tumor , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Pancreáticas/patología , Receptores de Estrógenos/biosíntesis , Receptores Acoplados a Proteínas G/biosíntesis , Sensibilidad y Especificidad
17.
Am J Obstet Gynecol ; 203(3): 242.e1-5, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20605134

RESUMEN

OBJECTIVE: We sought to evaluate the expression of G protein-coupled receptor 30 (GPR30) and estrogen receptor (ER)beta in uterine carcinosarcoma (CS). STUDY DESIGN: Immunohistochemistry was performed using antibodies to GPR30, ERbeta, ERalpha, and progesterone receptor (PR). The staining intensity and percentage of positive cells were scored for each tissue section. Expression levels were compared using the Wilcoxon rank sum test. Correlation was evaluated by Spearman rho and logistic regression. RESULTS: Compared with normal endometrium, CS had lower ERalpha and PR expression (both P < .01) but higher GPR30 epithelial expression (P = .03). Advanced-stage CS had higher GPR30 (P < .01) and ERbeta (P = .02) epithelial expression compared with early-stage CS. Expression of GPR30 and ERbeta correlated with each other (P < .01), and not with ERalpha or PR. CONCLUSION: In uterine CS, GPR30 and ERbeta are coordinately overexpressed and expression levels increase in advanced-stage disease, supporting the involvement of alternative ERs in disease progression.


Asunto(s)
Carcinosarcoma/metabolismo , Progresión de la Enfermedad , Receptor beta de Estrógeno/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Neoplasias Uterinas/metabolismo , Anciano , Carcinosarcoma/patología , Endometrio/metabolismo , Epitelio/metabolismo , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Inmunohistoquímica , Receptores de Estrógenos , Receptores de Progesterona/metabolismo , Neoplasias Uterinas/patología
18.
Clin Cancer Res ; 16(11): 2999-3010, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20404007

RESUMEN

PURPOSE: This study was undertaken to examine the role of the insulin-like growth factor (IGF) signaling pathway in the response of ovarian cancer cells to Taxol and to evaluate the significance of this pathway in human epithelial ovarian tumors. EXPERIMENTAL DESIGN: The effect of Taxol treatment on AKT activation in A2780 ovarian carcinoma cells was evaluated using antibodies specific for phospho-AKT. To study the drug-resistant phenotype, we developed a Taxol-resistant cell line, HEY-T30, derived from HEY ovarian carcinoma cells. IGF2 expression was measured by real-time PCR. A type 1 IGF receptor (IGF1R) inhibitor, NVP-AEW541, and IGF2 small interfering RNA were used to evaluate the effect of IGF pathway inhibition on proliferation and Taxol sensitivity. IGF2 protein expression was evaluated by immunohistochemistry in 115 epithelial ovarian tumors and analyzed in relation to clinical/pathologic factors using the chi(2) or Fisher's exact tests. The influence of IGF2 expression on survival was studied with Cox regression. RESULTS: Taxol-induced AKT phosphorylation required IGF1R tyrosine kinase activity and was associated with upregulation of IGF2. Resistant cells had higher IGF2 expression compared with sensitive cells, and IGF pathway inhibition restored sensitivity to Taxol. High IGF2 tumor expression correlated with advanced stage (P < 0.001) and tumor grade (P < 0.01) and reduced disease-free survival (P < 0.05). CONCLUSIONS: IGF2 modulates Taxol resistance, and tumor IGF2 expression is a candidate prognostic biomarker in epithelial ovarian tumors. IGF pathway inhibition sensitizes drug-resistant ovarian carcinoma cells to Taxol. Such novel findings suggest that IGF2 represents a therapeutic target in ovarian cancer, particularly in the setting of Taxol resistance.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Factor II del Crecimiento Similar a la Insulina/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Paclitaxel/uso terapéutico , Biomarcadores de Tumor/análisis , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia sin Enfermedad , Resistencia a Antineoplásicos , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Factor II del Crecimiento Similar a la Insulina/análisis , Neoplasias Ováricas/metabolismo , Fosforilación , Pronóstico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas/farmacología , Pirroles/farmacología , ARN Interferente Pequeño/farmacología , Receptor IGF Tipo 1/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
19.
Breast Cancer Res Treat ; 123(1): 51-8, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19902352

RESUMEN

GPR30 is a novel G protein-coupled estrogen receptor (ER) associated with metastases in breast cancer (BC) and poor survival in endometrial and ovarian tumors. The association of GPR30 expression with inflammatory breast cancer (IBC), an aggressive and commonly hormone-independent form of BC, has not been studied. GPR30, ER, progesterone receptor (PR), epidermal growth factor receptor (EGFR), and HER-2 expression were assessed by immunohistochemistry (and FISH for HER-2) in 88 primary IBCs. GPR30 expression was correlated with patient overall survival (OS), disease-free survival (DFS), pathologic variables, and other biomarkers. GPR30 expression was found in 69% of IBC cases. ER, PR, HER-2, and EGFR were found in 43, 35, 39, and 34% of IBC cases, respectively. GPR30 expression correlated inversely with ER expression (P = 0.02). Co-expression of ER and GPR30 was found in 24% of IBC samples; 19% expressed only ER and 46% expressed only GPR30. Univariate analysis showed no association between GPR30 expression and OS or DFS. However, co-expression of ER and GPR30 was associated with improved OS (P < 0.03) and marginally with DFS (P < 0.06); the absence of both ER and GPR30 was associated with worse OS and DFS (P = 0.03 for both). Multivariate analysis identified ER as an independent prognostic factor of OS (P = 0.008) and DFS (P = 0.02). The majority of IBC tumors are GPR30-positive, suggesting that estrogen signaling may be active in ER-negative IBC patients. These findings suggest potential new therapeutic targets for IBC such as novel endocrine agents or direct modulation of GPR30.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias de la Mama/metabolismo , Receptores de Estrógenos/biosíntesis , Receptores Acoplados a Proteínas G/biosíntesis , Adulto , Anciano , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Supervivencia sin Enfermedad , Receptores ErbB/biosíntesis , Femenino , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Estimación de Kaplan-Meier , Persona de Mediana Edad , Estadificación de Neoplasias , Modelos de Riesgos Proporcionales , Receptor ErbB-2/biosíntesis , Receptores de Progesterona/biosíntesis , Adulto Joven
20.
Genes Chromosomes Cancer ; 48(11): 983-92, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19681120

RESUMEN

Cervical cancer (CC) is the second most common cancer in women. Currently, no tractable molecular-based therapeutic targets exist for patients with invasive CC and no predictive markers of risk assessment for progression of precancerous lesions are identified. New molecular insights into CC pathogenesis are urgently needed. Towards this goal, we first determined the copy number alterations of chromosome 4 and then examined the role of PCDH10 mapped to 4q28 as a candidate tumor suppressor gene. We identified monosomy 4 in 47% of 81 invasive CC studied by SNP array and found that 91% of 130 invasive CC harboring methylation in the promoter region of the PCDH10 gene. We then showed that aberrant promoter hypermethylation of PCDH10 is associated with downregulation of gene expression and cell lines exposed to demethylating agent resulted in profound reactivated gene expression. We also showed that the promoter methylation in the PCDH10 gene occurs at an earliest identifiable stage of low-grade squamous intraepithelial lesion. Our studies demonstrate that inactivation of PCDH10 may be a critical event in CC progression and form a potentially useful therapeutic target for CC.


Asunto(s)
Cadherinas/genética , Metilación de ADN , Neoplasias del Cuello Uterino/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Cromosomas Humanos Par 4 , Variaciones en el Número de Copia de ADN , Análisis Mutacional de ADN , Progresión de la Enfermedad , Epigénesis Genética , Femenino , Dosificación de Gen , Expresión Génica , Humanos , Monosomía , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Protocadherinas , Neoplasias del Cuello Uterino/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA