Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(1)2022 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-36613564

RESUMEN

The search for immunotherapy biomarkers in Microsatellite Instability High/Deficient Mismatch Repair system (MSI-H/dMMR) metastatic colorectal cancer (mCRC) is an unmet need. Sixteen patients with mCRC and MSI-H/dMMR (determined by either immunohistochemistry or polymerase chain reaction) treated with PD-1/PD-L1 inhibitors at our institution were included. According to whether the progression-free survival with PD-1/PD-L1 inhibitors was longer than 6 months or shorter, patients were clustered into the IT-responder group (n: 9 patients) or IT-resistant group (n: 7 patients), respectively. In order to evaluate determinants of benefit with PD-1/PD-L1 inhibitors, we performed multimodal analysis including genomics (through NGS panel tumour-only with 431 genes) and the immune microenvironment (using CD3, CD8, FOXP3 and PD-L1 antibodies). The following mutations were more frequent in IT-resistant compared with IT-responder groups: B2M (4/7 versus 2/9), CTNNB1 (2/7 versus 0/9), and biallelic PTEN (3/7 versus 1/9). Biallelic ARID1A mutations were found exclusively in the IT-responder group (4/9 patients). Tumour mutational burden did not correlate with immunotherapy benefit, neither the rate of indels in homopolymeric regions. Of note, biallelic ARID1A mutated tumours had the highest immune infiltration and PD-L1 scores, contrary to tumours with CTNNB1 mutation. Immune microenvironment analysis showed higher densities of different T cell subpopulations and PD-L1 expression in IT-responders. Misdiagnosis of MSI-H/dMMR inferred by discordances between immunohistochemistry and polymerase chain reaction was only found in the IT-resistant population (3/7 patients). Biallelic ARID1A mutations and Wnt signalling activation through CTNNB1 mutation were associated with high and low T cell immune infiltrates, respectively, and deserve special attention as determinants of response to PD-1/PD-L1 inhibitors. The non-MSI-H phenotype in dMMR is associated with poor benefit to immunotherapy. Our results suggest that mechanisms of resistance to immunotherapy are multi-factorial.


Asunto(s)
Neoplasias del Colon , Neoplasias Colorrectales , Humanos , Antígeno B7-H1/genética , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Reparación de la Incompatibilidad de ADN , Receptor de Muerte Celular Programada 1/genética , Neoplasias del Colon/genética , Neoplasias Colorrectales/terapia , Neoplasias Colorrectales/tratamiento farmacológico , Repeticiones de Microsatélite , Inestabilidad de Microsatélites , Inmunoterapia , Microambiente Tumoral/genética
2.
Radiology ; 299(1): 109-119, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33497314

RESUMEN

Background Reliable predictive imaging markers of response to immune checkpoint inhibitors are needed. Purpose To develop and validate a pretreatment CT-based radiomics signature to predict response to immune checkpoint inhibitors in advanced solid tumors. Materials and Methods In this retrospective study, a radiomics signature was developed in patients with advanced solid tumors (including breast, cervix, gastrointestinal) treated with anti-programmed cell death-1 or programmed cell death ligand-1 monotherapy from August 2012 to May 2018 (cohort 1). This was tested in patients with bladder and lung cancer (cohorts 2 and 3). Radiomics variables were extracted from all metastases delineated at pretreatment CT and selected by using an elastic-net model. A regression model combined radiomics and clinical variables with response as the end point. Biologic validation of the radiomics score with RNA profiling of cytotoxic cells (cohort 4) was assessed with Mann-Whitney analysis. Results The radiomics signature was developed in 85 patients (cohort 1: mean age, 58 years ± 13 [standard deviation]; 43 men) and tested on 46 patients (cohort 2: mean age, 70 years ± 12; 37 men) and 47 patients (cohort 3: mean age, 64 years ± 11; 40 men). Biologic validation was performed in a further cohort of 20 patients (cohort 4: mean age, 60 years ± 13; 14 men). The radiomics signature was associated with clinical response to immune checkpoint inhibitors (area under the curve [AUC], 0.70; 95% CI: 0.64, 0.77; P < .001). In cohorts 2 and 3, the AUC was 0.67 (95% CI: 0.58, 0.76) and 0.67 (95% CI: 0.56, 0.77; P < .001), respectively. A radiomics-clinical signature (including baseline albumin level and lymphocyte count) improved on radiomics-only performance (AUC, 0.74 [95% CI: 0.63, 0.84; P < .001]; Akaike information criterion, 107.00 and 109.90, respectively). Conclusion A pretreatment CT-based radiomics signature is associated with response to immune checkpoint inhibitors, likely reflecting the tumor immunophenotype. © RSNA, 2021 Online supplemental material is available for this article. See also the editorial by Summers in this issue.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Tomografía Computarizada por Rayos X/métodos , Anciano , Biomarcadores de Tumor , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
3.
Oncogene ; 42(35): 2610-2628, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37468678

RESUMEN

Epithelial/Mesenchymal (E/M) plasticity plays a fundamental role both in embryogenesis and during tumorigenesis. The receptor for advanced glycation end products (RAGE) is a driver of cell plasticity in fibrotic diseases; however, its role and molecular mechanism in triple-negative breast cancer (TNBC) remains unclear. Here, we demonstrate that RAGE signaling maintains the mesenchymal phenotype of aggressive TNBC cells by enforcing the expression of SNAIL1. Besides, we uncover a crosstalk mechanism between the TGF-ß and RAGE pathways that is required for the acquisition of mesenchymal traits in TNBC cells. Consistently, RAGE inhibition elicits epithelial features that block migration and invasion capacities. Next, since RAGE is a sensor of the tumor microenvironment, we modeled acute acidosis in TNBC cells and showed it promotes enhanced production of RAGE ligands and the activation of RAGE-dependent invasive properties. Furthermore, acute acidosis increases SNAIL1 levels and tumor cell invasion in a RAGE-dependent manner. Finally, we demonstrate that in vivo inhibition of RAGE reduces metastasis incidence and expands survival, consistent with molecular effects that support the relevance of RAGE signaling in E/M plasticity. These results uncover new molecular insights on the regulation of E/M phenotypes in cancer metastasis and provide rationale for pharmacological intervention of this signaling axis.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/patología , Receptor para Productos Finales de Glicación Avanzada/genética , Línea Celular Tumoral , Transducción de Señal , Fenotipo , Transición Epitelial-Mesenquimal , Movimiento Celular , Microambiente Tumoral
4.
Cancers (Basel) ; 15(14)2023 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-37509274

RESUMEN

MPM is an aggressive disease with an immunosuppressive tumor microenvironment, and interest in exploring immunotherapy in this disease has been increasing. In the first line of treatment, the combination of nivolumab and ipilimumab demonstrated an improvement in survival over chemotherapy. The presence of TILs has been recognized as a marker of antitumor immune response to chemotherapy in solid tumors. The aim of our study is to identify the effect of treatment on immune cells and the immune gene profile in MPM. We investigated the changes in expression of TILs in 10 human MPM paired tumor tissues using immunohistochemistry and gene expression analysis from paired untreated and treated samples. In this small series, we demonstrated that during the evolution of disease without any treatment there was an increase in the inflammatory component in tumor samples. After systemic treatment there was a decrease in the number of TILs. We observed that after systemic treatment or disease progression immune gene signatures were suppressed. Our integrated analysis of paired samples with immune profile and genomic changes on MPM suggested that during the evolution of the disease the immune system tends to switch, turning off with treatment.

5.
Med ; 4(10): 710-727.e5, 2023 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-37572657

RESUMEN

BACKGROUND: Immunotherapy is effective, but current biomarkers for patient selection have proven modest sensitivity. Here, we developed VIGex, an optimized gene signature based on the expression level of 12 genes involved in immune response with RNA sequencing. METHODS: We implemented VIGex using the nCounter platform (Nanostring) on a large clinical cohort encompassing 909 tumor samples across 45 tumor types. VIGex was developed as a continuous variable, with cutoffs selected to detect three main categories (hot, intermediate-cold and cold) based on the different inflammatory status of the tumor microenvironment. FINDINGS: Hot tumors had the highest VIGex scores and exhibited an increased abundance of tumor-infiltrating lymphocytes as compared with the intermediate-cold and cold. VIGex scores varied depending on tumor origin and anatomic site of metastases, with liver metastases showing an immunosuppressive tumor microenvironment. The predictive power of VIGex-Hot was observed in a cohort of 98 refractory solid tumor from patients treated in early-phase immunotherapy trials and its clinical performance was confirmed through an extensive metanalysis across 13 clinically annotated gene expression datasets from 877 patients treated with immunotherapy agents. Last, we generated a pan-cancer biomarker platform that integrates VIGex categories with the expression levels of immunotherapy targets under development in early-phase clinical trials. CONCLUSIONS: Our results support the clinical utility of VIGex as a tool to aid clinicians for patient selection and personalized immunotherapy interventions. FUNDING: BBVA Foundation; 202-2021 Division of Medical Oncology and Hematology Fellowship award; Princess Margaret Cancer Center.


Asunto(s)
Neoplasias , Humanos , Neoplasias/genética , Neoplasias/terapia , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/metabolismo , Factores Inmunológicos/metabolismo , Factores Inmunológicos/uso terapéutico , Oncología Médica , Microambiente Tumoral/genética
6.
Nat Med ; 28(10): 2162-2170, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36097219

RESUMEN

Anti-BRAF/EGFR therapy was recently approved for the treatment of metastatic BRAFV600E colorectal cancer (mCRCBRAF-V600E). However, a large fraction of patients do not respond, underscoring the need to identify molecular determinants of treatment response. Using whole-exome sequencing in a discovery cohort of patients with mCRCBRAF-V600E treated with anti-BRAF/EGFR therapy, we found that inactivating mutations in RNF43, a negative regulator of WNT, predict improved response rates and survival outcomes in patients with microsatellite-stable (MSS) tumors. Analysis of an independent validation cohort confirmed the relevance of RNF43 mutations to predicting clinical benefit (72.7% versus 30.8%; P = 0.03), as well as longer progression-free survival (hazard ratio (HR), 0.30; 95% confidence interval (CI), 0.12-0.75; P = 0.01) and overall survival (HR, 0.26; 95% CI, 0.10-0.71; P = 0.008), in patients with MSS-RNF43mutated versus MSS-RNF43wild-type tumors. Microsatellite-instable tumors invariably carried a wild-type-like RNF43 genotype encoding p.G659fs and presented an intermediate response profile. We found no association of RNF43 mutations with patient outcomes in a control cohort of patients with MSS-mCRCBRAF-V600E tumors not exposed to anti-BRAF targeted therapies. Overall, our findings suggest a cross-talk between the MAPK and WNT pathways that may modulate the antitumor activity of anti-BRAF/EGFR therapy and uncover predictive biomarkers to optimize the clinical management of these patients.


Asunto(s)
Neoplasias Colorrectales , Ubiquitina-Proteína Ligasas , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Receptores ErbB/genética , Humanos , Inestabilidad de Microsatélites , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Ubiquitina-Proteína Ligasas/genética
7.
Cancers (Basel) ; 13(21)2021 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-34771584

RESUMEN

Formalin-fixed, paraffin-embedded (FFPE) tissues represent the most widely available clinical material to study colorectal cancer (CRC). However, the accuracy and clinical validity of FFPE microbiome profiling in CRC is uncertain. Here, we compared the microbial composition of 10 paired fresh-frozen (FF) and FFPE CRC tissues using 16S rRNA sequencing and RNA-ISH. Both sample types showed different microbial diversity and composition. FF samples were enriched in archaea and representative CRC-associated bacteria, such as Firmicutes, Bacteroidetes and Fusobacteria. Conversely, FFPE samples were mainly enriched in typical contaminants, such as Sphingomonadales and Rhodobacterales. RNA-ISH in FFPE tissues confirmed the presence of CRC-associated bacteria, such as Fusobacterium and Bacteroides, as well as Propionibacterium allowing discrimination between tumor-associated and contaminant taxa. An internal quality index showed that the degree of similarity within sample pairs inversely correlated with the dominance of contaminant taxa. Given the importance of FFPE specimens for larger studies in human cancer genomics, our findings may provide useful indications on potential confounding factors to consider for accurate and reproducible metagenomics analyses.

8.
NPJ Breast Cancer ; 7(1): 1, 2021 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-33397968

RESUMEN

Novel antibody-drug conjugates against HER2 are showing high activity in HER2-negative breast cancer (BC) with low HER2 expression (i.e., 1+ or 2+ and lack of ERBB2 amplification). However, the clinical and molecular features of HER2-low BC are yet to be elucidated. Here, we collected retrospective clinicopathological and PAM50 data from 3,689 patients with HER2-negative disease and made the following observations. First, the proportion of HER2-low was higher in HR-positive disease (65.4%) than triple-negative BC (TNBC, 36.6%). Second, within HR-positive disease, ERBB2 and luminal-related genes were more expressed in HER2-low than HER2 0. In contrast, no gene was found differentially expressed in TNBC according to HER2 expression. Third, within HER2-low, ERBB2 levels were higher in HR-positive disease than TNBC. Fourth, HER2-low was not associated with overall survival in HR-positive disease and TNBC. Finally, the reproducibility of HER2-low among pathologists was suboptimal. This study emphasizes the large biological heterogeneity of HER2-low BC, and the need to implement reproducible and sensitive assays to measure low HER2 expression.

9.
NPJ Breast Cancer ; 7(1): 73, 2021 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099718

RESUMEN

The biology of breast cancer response to neoadjuvant therapy is underrepresented in the literature and provides a window-of-opportunity to explore the genomic and microenvironment modulation of tumours exposed to therapy. Here, we characterised the mutational, gene expression, pathway enrichment and tumour-infiltrating lymphocytes (TILs) dynamics across different timepoints of 35 HER2-negative primary breast cancer patients receiving neoadjuvant eribulin therapy (SOLTI-1007 NEOERIBULIN-NCT01669252). Whole-exome data (N = 88 samples) generated mutational profiles and candidate neoantigens and were analysed along with RNA-Nanostring 545-gene expression (N = 96 samples) and stromal TILs (N = 105 samples). Tumour mutation burden varied across patients at baseline but not across the sampling timepoints for each patient. Mutational signatures were not always conserved across tumours. There was a trend towards higher odds of response and less hazard to relapse when the percentage of subclonal mutations was low, suggesting that more homogenous tumours might have better responses to neoadjuvant therapy. Few driver mutations (5.1%) generated putative neoantigens. Mutation and neoantigen load were positively correlated (R2 = 0.94, p = <0.001); neoantigen load was weakly correlated with stromal TILs (R2 = 0.16, p = 0.02). An enrichment in pathways linked to immune infiltration and reduced programmed cell death expression were seen after 12 weeks of eribulin in good responders. VEGF was downregulated over time in the good responder group and FABP5, an inductor of epithelial mesenchymal transition (EMT), was upregulated in cases that recurred (p < 0.05). Mutational heterogeneity, subclonal architecture and the improvement of immune microenvironment along with remodelling of hypoxia and EMT may influence the response to neoadjuvant treatment.

10.
Breast ; 53: 102-110, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32707454

RESUMEN

OBJECTIVE: Ki67 is a prognostic and predictive marker in breast cancer (BC). However, manual scoring (MS) by visual assessment suffers from high inter-observer variability which limits its clinical use. Here, we developed a new digital image analysis (DIA) workflow, named KiQuant for automated scoring of Ki67 and investigated its equivalence with standard pathologist's assessment. METHODS: Sequential immunohistochemistry of Ki67 and cytokeratin, for precise tumor cell recognition, were performed in the same section of 5 tissue microarrays containing 329 tumor cores from different breast cancer subtypes. Slides were digitalized and subjected to DIA and MS for Ki67 assessment. The intraclass correlation coefficient (ICC) and Bland-Altman plot were used to evaluate inter-observer reproducibility. The Kaplan-Meier analysis was used to determine the prognostic potential. RESULTS: KiQuant showed an excellent correlation with MS (ICC:0.905,95%CI:0.878-0.926) with satisfactory inter-run (ICC:0.917,95%CI:0.884-0.942) and inter-antibody reproducibilities (ICC:0.886,95%CI:0.820-0.929). The distance between KiQuant and MS increased with the magnitude of Ki67 measurement and positively correlated with analyzed tumor area and breast cancer subtype. Agreement rates between KiQuant and MS within the clinically relevant 14% and 30% cut-off points ranged from 33% to 44% with modest interobserver reproducibility below the 20% cut-off (0.606, 95%CI:0.467-0.727). High Ki67 by KiQuant correlated with worse outcome in all BC and in the luminal subtype (P = 0.028 and P = 0.043, respectively). For MS, the association with survival was significant only in 1 out of 3 observers. CONCLUSIONS: KiQuant represents an easy and accurate methodology for Ki67 measurement providing a step toward utilizing Ki67 in the clinical setting.


Asunto(s)
Neoplasias de la Mama/patología , Procesamiento de Imagen Asistido por Computador/métodos , Inmunohistoquímica/métodos , Antígeno Ki-67/análisis , Programas Informáticos , Biomarcadores de Tumor/análisis , Femenino , Humanos , Estimación de Kaplan-Meier , Queratinas/análisis , Variaciones Dependientes del Observador , Pronóstico , Reproducibilidad de los Resultados
11.
Artículo en Inglés | MEDLINE | ID: mdl-32923891

RESUMEN

Most academic precision oncology programs have been designed to facilitate enrollment of patients in early clinical trials with matched targeted agents. Over the last decade, major changes were seen both in the targetable molecular alteration landscape and in drug development trends. In this article, we describe how the Vall d'Hebron Institute of Oncology molecular prescreening program adapted to a dynamic model of biomarker-drug codevelopment. We started with a tumor-agnostic hotspot mutation panel plus in situ hybridization and immunohistochemistry of selected markers and subsequently transitioned to tumor-specific amplicon-based next-generation sequencing (NGS) tests together with custom copy number, fusion, and outlier gene expression panels. All assays are optimized for archived formalin-fixed paraffin-embedded tumor tissues without matched germline sequencing. In parallel, biomarker-matched trials evolved from a scenario of few targets and large populations (such as PI3K inhibitors in PIK3CA mutants) to a complex situation with many targets and small populations (such as multiple targetable fusion events). Recruitment rates in clinical trials with mandatory biomarkers decreased over the last 3 years. Molecular tumor board meetings proved critical to guide oncologists on emerging biomarkers for clinical testing and interpretation of NGS results. The substantial increase of immunotherapy trials had a major impact in target prioritization and guided clinical implementation of new markers, such as tumor mutational burden, with larger exon-based NGS assays and gene expression signatures to capture microenvironment infiltration patterns. This new multiomics era of precision oncology is expected to increase the opportunities for early clinical trial matching.

12.
J Natl Cancer Inst ; 112(1): 46-54, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31037288

RESUMEN

BACKGROUND: Identification of HER2-positive breast cancers with high anti-HER2 sensitivity could help de-escalate chemotherapy. Here, we tested a clinically applicable RNA-based assay that combines ERBB2 and the HER2-enriched (HER2-E) intrinsic subtype in HER2-positive disease treated with dual HER2-blockade without chemotherapy. METHODS: A research-based PAM50 assay was applied in 422 HER2-positive tumors from five II-III clinical trials (SOLTI-PAMELA, TBCRC023, TBCRC006, PER-ELISA, EGF104090). In SOLTI-PAMELA, TBCRC023, TBCRC006, and PER-ELISA, all patients had early disease and were treated with neoadjuvant lapatinib or pertuzumab plus trastuzumab for 12-24 weeks. Primary outcome was pathological complete response (pCR). In EGF104900, 296 women with advanced disease were randomized to receive either lapatinib alone or lapatinib plus trastuzumab. Progression-free survival (PFS), overall response rate (ORR), and overall survival (OS) were evaluated. RESULTS: A total of 305 patients with early and 117 patients with advanced HER2-positive disease were analyzed. In early disease, HER2-E represented 83.8% and 44.7% of ERBB2-high and ERBB2-low tumors, respectively. Following lapatinib and trastuzumab, the HER2-E and ERBB2 (HER2-E/ERBB2)-high group showed a higher pCR rate compared to the rest (44.5%, 95% confidence interval [CI] = 35.4% to 53.9% vs 11.6%, 95% CI = 6.9% to 18.0%; adjusted odds ratio [OR] = 6.05, 95% CI = 3.10 to 11.80, P < .001). Similar findings were observed with neoadjuvant trastuzumab and pertuzumab (pCR rate of 66.7% in HER2-E/ERBB2-high, 95% CI = 22.3% to 95.7% vs 14.7% in others, 95% CI = 4.9% to 31.1%; adjusted OR = 11.60, 95% CI = 1.66 to 81.10, P = .01). In the advanced setting, the HER2-E/ERBB2-high group was independently associated with longer PFS (hazard ratio [HR] = 0.52, 95% CI = 0.35 to 0.79, P < .001); higher ORR (16.3%, 95% CI = 8.9% to 26.2% vs 3.7%, 95% CI = 0.8% to 10.3%, P = .02); and longer OS (HR = 0.66, 95% CI = 0.44 to 0.97, P = .01). CONCLUSIONS: Combining HER2-E subtype and ERBB2 mRNA into a single assay identifies tumors with high responsiveness to HER2-targeted therapy. This biomarker could help de-escalate chemotherapy in approximately 40% of patients with HER2-positive breast cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Biomarcadores de Tumor , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Expresión Génica , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/genética , Adulto , Anciano , Anticuerpos Monoclonales Humanizados , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Femenino , Humanos , Lapatinib , Persona de Mediana Edad , Terapia Molecular Dirigida/métodos , Terapia Neoadyuvante , Estadificación de Neoplasias , Pronóstico , Reproducibilidad de los Resultados , Análisis de Supervivencia , Resultado del Tratamiento
13.
Tumori ; 95(4): 532-4, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19856670

RESUMEN

Imaging evaluation of uterine masses is important to assess the type of lesion and to target surgery, if surgical excision is necessary. This can be decisive in fertile women with benign masses resembling malignancies, in order to avoid overtreatment. In this study, the magnetic resonance imaging (MRI) appearance of cotyledonoid dissecting leiomyoma of the uterus, a rare benign variant of leiomyoma mimicking malignancy, is presented.


Asunto(s)
Leiomioma/patología , Imagen por Resonancia Magnética , Neoplasias Uterinas/patología , Adulto , Diagnóstico Diferencial , Femenino , Humanos
14.
Sci Rep ; 9(1): 13568, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31537838

RESUMEN

Protein biomarkers are widely used in cancer diagnosis, prognosis, and prediction of treatment response. Here we introduce the use of targeted multiplex proteomics (TMP) as a tool to simultaneously measure a panel of 54 proteins involved in oncogenic, tumour suppression, drug metabolism and resistance, in patients with metastatic colorectal cancer (mCRC). TMP provided valuable diagnostic information by unmasking an occult neuroendocrine differentiation and identifying a misclassified case based on abnormal proteins phenotype. No significant differences in protein levels between unpaired primary and metastatic samples were observed. Four proteins were found differentially expressed in KRAS-mutant as compared to wild-type tumours (overexpressed in mutant: KRAS, EGFR; overexpressed in wild-type: TOPO1, TOP2A). Survival analyses revealed the association between mesothelin expression and poor overall survival, whereas lack of PTEN protein expression associated with lower progression-free survival with anti-EGFR-based therapy in the first-line setting for patients with RAS wild-type tumour. Finally, outlier analysis identified putative targetable proteins in 65% of patients lacking a targetable genomic alteration. Our data show that TMP constitutes a promising, novel molecular prescreening tool in mCRC to identify protein expression alterations that may impact on patient outcomes and more precisely guide patient eligibility to clinical trials with novel targeted experimental therapies.


Asunto(s)
Biomarcadores/metabolismo , Neoplasias Colorrectales/patología , Proteínas Ligadas a GPI/metabolismo , Fosfohidrolasa PTEN/metabolismo , Proteómica/métodos , Adulto , Anciano , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/mortalidad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Mesotelina , Persona de Mediana Edad , Mutación , Metástasis de la Neoplasia , Pronóstico , Proteínas Proto-Oncogénicas p21(ras)/genética , Análisis de Supervivencia
16.
Sci Transl Med ; 10(461)2018 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-30282693

RESUMEN

T cell bispecific antibodies (TCBs) are engineered molecules that include, within a single entity, binding sites to the T cell receptor and to tumor-associated or tumor-specific antigens. The receptor tyrosine kinase HER2 is a tumor-associated antigen in ~25% of breast cancers. TCBs targeting HER2 may result in severe toxicities, likely due to the expression of HER2 in normal epithelia. About 40% of HER2-positive tumors express p95HER2, a carboxyl-terminal fragment of HER2. Using specific antibodies, here, we show that p95HER2 is not expressed in normal tissues. We describe the development of p95HER2-TCB and show that it has a potent antitumor effect on p95HER2-expressing breast primary cancers and brain lesions. In contrast with a TCB targeting HER2, p95HER2-TCB has no effect on nontransformed cells that do not overexpress HER2. These data pave the way for the safe treatment of a subgroup of HER2-positive tumors by targeting a tumor-specific antigen.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/inmunología , Receptor ErbB-2/inmunología , Linfocitos T/inmunología , Animales , Neoplasias de la Mama/patología , Complejo CD3/inmunología , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Science ; 358(6369): 1443-1448, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29170280

RESUMEN

Colorectal cancers comprise a complex mixture of malignant cells, nontransformed cells, and microorganisms. Fusobacterium nucleatum is among the most prevalent bacterial species in colorectal cancer tissues. Here we show that colonization of human colorectal cancers with Fusobacterium and its associated microbiome-including Bacteroides, Selenomonas, and Prevotella species-is maintained in distal metastases, demonstrating microbiome stability between paired primary and metastatic tumors. In situ hybridization analysis revealed that Fusobacterium is predominantly associated with cancer cells in the metastatic lesions. Mouse xenografts of human primary colorectal adenocarcinomas were found to retain viable Fusobacterium and its associated microbiome through successive passages. Treatment of mice bearing a colon cancer xenograft with the antibiotic metronidazole reduced Fusobacterium load, cancer cell proliferation, and overall tumor growth. These observations argue for further investigation of antimicrobial interventions as a potential treatment for patients with Fusobacterium-associated colorectal cancer.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/microbiología , Antibacterianos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/microbiología , Fusobacterium/efectos de los fármacos , Metronidazol/farmacología , Microbiota/efectos de los fármacos , Adenocarcinoma/secundario , Animales , Antibacterianos/farmacología , Bacteroides/efectos de los fármacos , Carcinogénesis , Neoplasias Colorrectales/patología , Fusobacterium/genética , Fusobacterium/aislamiento & purificación , Células HT29 , Humanos , Neoplasias Hepáticas/microbiología , Neoplasias Hepáticas/secundario , Metronidazol/uso terapéutico , Ratones , Prevotella/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Hum Pathol ; 34(7): 639-45, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12874758

RESUMEN

Amplification and/or overexpression of HER2/neu have been documented in many types of epithelial tumor and recently has been reported in sarcomas, particularly in osteosarcomas. But the role of HER2/neu alterations in soft tissue tumors remains poorly understood. Thus the present study investigates the expression of HER2/neu in 13 patients with synovial sarcoma (SS). In this study, HER2/neu mRNA levels were measured in frozen tissue samples using a real-time reverse transcription-polymerase chain reaction assay; protein expression was assessed by immunohistochemistry using an anti-HER2/neu polyclonal antibody. Six normal skeletal muscle specimens were used to establish basal levels of HER2/neu mRNA. HER2/neu transcripts were detected in all normal tissues and SSs. Four of 13 sarcomas (31%) demonstrated HER2/neu mRNA levels above the mean value, whereas 3 tumors (23%) displayed HER2/neu protein overexpression. Both membranous and cytoplasmic patterns of immunostaining were observed, and a strong correlation was found between protein expression and mRNA level (P = 0.01). Increased HER2/neu mRNA levels were significantly associated with a lower risk of developing recurrences (P = 0.02). Moreover, none of the patients with HER2/neu overexpression developed metastasis. Our data demonstrate that HER2/neu is expressed in SSs and that both membrane and cytoplasmic HER2/neu expression correlate with mRNA levels. Our results show that the presence of increased levels of HER2/neu in SSs is associated with a more favorable clinical course. Further studies are needed to assess the role of this oncogene in SSs and to evaluate the application of inhibitory humanized monoclonal antibodies in the treatment regimens for this malignancy.


Asunto(s)
Genes erbB-2 , Receptor ErbB-2/genética , Sarcoma Sinovial/genética , Neoplasias de los Tejidos Blandos/genética , Adolescente , Adulto , Anciano , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Pronóstico , ARN Mensajero/genética , ARN Neoplásico/análisis , Receptor ErbB-2/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcoma Sinovial/metabolismo , Sarcoma Sinovial/secundario , Neoplasias de los Tejidos Blandos/metabolismo , Neoplasias de los Tejidos Blandos/patología
19.
Cancer Res ; 68(4): 1110-8, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18281486

RESUMEN

Epithelial ovarian carcinoma (EOC) arises from the ovarian surface epithelium (OSE), a monolayer of poorly differentiated epithelial cells that lines the ovary. The molecular mechanisms underlying EOC invasion into the surrounding stroma and dissemination to the peritoneum and to retroperitoneal lymph nodes are still unclear. Here, we analyzed the expression and the functional role of the cell adhesion molecule L1 during EOC development. In patient-derived samples, L1 was expressed both in OSE and in a subset of EOC, in the latter being mostly restricted to the invasive areas of the tumors. The expression of L1 correlated significantly with poor outcome and with unfavorable clinicopathologic features of the disease. The peculiar expression pattern of L1 in normal OSE and invasive EOC raised the possibility that this adhesion molecule serves a different function in nontransformed versus neoplastic ovarian epithelial cells. Indeed, we showed that in OSE cells L1 supports cell-cell adhesion and enhances apoptosis, whereas it has no effect on cell proliferation and invasion. In contrast, L1 inhibits cell-cell adhesion and apoptosis in ovarian carcinoma cells, where it promotes malignancy-related properties, such as cell proliferation, Erk1/2-dependent and phosphoinositide 3-kinase-dependent invasion, and transendothelial migration. Interestingly, a crosstalk with the fibroblast growth factor receptor signaling is implicated in the promalignant function of L1 in tumor cells. Our findings point to L1 as an EOC biomarker correlating with poor prognosis, and highlight a switch in L1 function associated to the neoplastic transformation of ovarian epithelial cells, thus implicating L1 as a potential therapeutic target.


Asunto(s)
Molécula L1 de Adhesión de Célula Nerviosa/biosíntesis , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Ovario/metabolismo , Apoptosis/fisiología , Adhesión Celular/fisiología , Comunicación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Endoteliales/patología , Células Epiteliales/metabolismo , Matriz Extracelular/metabolismo , Femenino , Humanos , Invasividad Neoplásica , Molécula L1 de Adhesión de Célula Nerviosa/genética , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Neoplasias Ováricas/genética , Ovario/citología , ARN Interferente Pequeño/genética , Receptor Cross-Talk , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Transfección
20.
Breast Cancer Res Treat ; 101(3): 349-53, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17009109

RESUMEN

BACKGROUND: Metaplastic carcinoma of the breast is a rare form of breast cancer and has an uncertain prognostic significance. The purpose of the present study was to compare the clinical course, and prognosis, between this type of tumor and poorly differentiated ductal carcinoma. PATIENTS AND METHODS: We analyzed 37 cases of metaplastic carcinoma of the breast treated at our institution (European Institute of Oncology in Milan, Italy) between 1997 and 2004, comparing them with 72 cases (control group) of poorly differentiated ductal carcinoma. All 109 patients had negative receptors and were G3 at final histology. The control cases were matched according to year of surgery, pT (pT1 vs. pT2/3/4), and pN (absent vs. present). RESULTS: Of the 37 patients, eleven died from disease progression, eight developed metastatic disease and two experienced local recurrence. In the control group (72 patients) we observed three deaths due to disease progression, 13 distant metastases, and two local recurrences. CONCLUSION: The overall survival in the metaplastic carcinoma group was significantly worse than in the control group. As regards to disease-free survival, there was no statistically significant difference between the two groups.


Asunto(s)
Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Carcinoma Intraductal no Infiltrante/mortalidad , Carcinoma Intraductal no Infiltrante/patología , Carcinoma Lobular/mortalidad , Carcinoma Lobular/patología , Recurrencia Local de Neoplasia/mortalidad , Neoplasias de la Mama/secundario , Neoplasias de la Mama/terapia , Carcinoma Intraductal no Infiltrante/secundario , Carcinoma Intraductal no Infiltrante/terapia , Carcinoma Lobular/secundario , Carcinoma Lobular/terapia , Estudios de Seguimiento , Humanos , Mastectomía , Metaplasia/mortalidad , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/radioterapia , Recurrencia Local de Neoplasia/cirugía , Estadificación de Neoplasias , Neoplasias Primarias Secundarias/mortalidad , Neoplasias Primarias Secundarias/patología , Neoplasias Primarias Secundarias/radioterapia , Neoplasias Primarias Secundarias/cirugía , Pronóstico , Análisis de Supervivencia , Resultado del Tratamiento , Población Blanca
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA