Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
2.
BMC Med ; 22(1): 9, 2024 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-38191387

RESUMEN

BACKGROUND: Due to the abundant usage of chemotherapy in young triple-negative breast cancer (TNBC) patients, the unbiased prognostic value of BRCA1-related biomarkers in this population remains unclear. In addition, whether BRCA1-related biomarkers modify the well-established prognostic value of stromal tumor-infiltrating lymphocytes (sTILs) is unknown. This study aimed to compare the outcomes of young, node-negative, chemotherapy-naïve TNBC patients according to BRCA1 status, taking sTILs into account. METHODS: We included 485 Dutch women diagnosed with node-negative TNBC under age 40 between 1989 and 2000. During this period, these women were considered low-risk and did not receive chemotherapy. BRCA1 status, including pathogenic germline BRCA1 mutation (gBRCA1m), somatic BRCA1 mutation (sBRCA1m), and tumor BRCA1 promoter methylation (BRCA1-PM), was assessed using DNA from formalin-fixed paraffin-embedded tissue. sTILs were assessed according to the international guideline. Patients' outcomes were compared using Cox regression and competing risk models. RESULTS: Among the 399 patients with BRCA1 status, 26.3% had a gBRCA1m, 5.3% had a sBRCA1m, 36.6% had tumor BRCA1-PM, and 31.8% had BRCA1-non-altered tumors. Compared to BRCA1-non-alteration, gBRCA1m was associated with worse overall survival (OS) from the fourth year after diagnosis (adjusted HR, 2.11; 95% CI, 1.18-3.75), and this association attenuated after adjustment for second primary tumors. Every 10% sTIL increment was associated with 16% higher OS (adjusted HR, 0.84; 95% CI, 0.78-0.90) in gBRCA1m, sBRCA1m, or BRCA1-non-altered patients and 31% higher OS in tumor BRCA1-PM patients. Among the 66 patients with tumor BRCA1-PM and ≥ 50% sTILs, we observed excellent 15-year OS (97.0%; 95% CI, 92.9-100%). Conversely, among the 61 patients with gBRCA1m and < 50% sTILs, we observed poor 15-year OS (50.8%; 95% CI, 39.7-65.0%). Furthermore, gBRCA1m was associated with higher (adjusted subdistribution HR, 4.04; 95% CI, 2.29-7.13) and tumor BRCA1-PM with lower (adjusted subdistribution HR, 0.42; 95% CI, 0.19-0.95) incidence of second primary tumors, compared to BRCA1-non-alteration. CONCLUSIONS: Although both gBRCA1m and tumor BRCA1-PM alter BRCA1 gene transcription, they are associated with different outcomes in young, node-negative, chemotherapy-naïve TNBC patients. By combining sTILs and BRCA1 status for risk classification, we were able to identify potential subgroups in this population to intensify and optimize adjuvant treatment.


Asunto(s)
Neoplasias Primarias Secundarias , Neoplasias de la Mama Triple Negativas , Humanos , Femenino , Adulto , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Adyuvantes Inmunológicos , Etnicidad , Biomarcadores , Proteína BRCA1/genética
3.
Mod Pathol ; 37(4): 100439, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38286221

RESUMEN

This work puts forth and demonstrates the utility of a reporting framework for collecting and evaluating annotations of medical images used for training and testing artificial intelligence (AI) models in assisting detection and diagnosis. AI has unique reporting requirements, as shown by the AI extensions to the Consolidated Standards of Reporting Trials (CONSORT) and Standard Protocol Items: Recommendations for Interventional Trials (SPIRIT) checklists and the proposed AI extensions to the Standards for Reporting Diagnostic Accuracy (STARD) and Transparent Reporting of a Multivariable Prediction model for Individual Prognosis or Diagnosis (TRIPOD) checklists. AI for detection and/or diagnostic image analysis requires complete, reproducible, and transparent reporting of the annotations and metadata used in training and testing data sets. In an earlier work by other researchers, an annotation workflow and quality checklist for computational pathology annotations were proposed. In this manuscript, we operationalize this workflow into an evaluable quality checklist that applies to any reader-interpreted medical images, and we demonstrate its use for an annotation effort in digital pathology. We refer to this quality framework as the Collection and Evaluation of Annotations for Reproducible Reporting of Artificial Intelligence (CLEARR-AI).


Asunto(s)
Inteligencia Artificial , Lista de Verificación , Humanos , Pronóstico , Procesamiento de Imagen Asistido por Computador , Proyectos de Investigación
4.
Histopathology ; 84(6): 915-923, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38433289

RESUMEN

A growing body of research supports stromal tumour-infiltrating lymphocyte (TIL) density in breast cancer to be a robust prognostic and predicive biomarker. The gold standard for stromal TIL density quantitation in breast cancer is pathologist visual assessment using haematoxylin and eosin-stained slides. Artificial intelligence/machine-learning algorithms are in development to automate the stromal TIL scoring process, and must be validated against a reference standard such as pathologist visual assessment. Visual TIL assessment may suffer from significant interobserver variability. To improve interobserver agreement, regulatory science experts at the US Food and Drug Administration partnered with academic pathologists internationally to create a freely available online continuing medical education (CME) course to train pathologists in assessing breast cancer stromal TILs using an interactive format with expert commentary. Here we describe and provide a user guide to this CME course, whose content was designed to improve pathologist accuracy in scoring breast cancer TILs. We also suggest subsequent steps to translate knowledge into clinical practice with proficiency testing.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Patólogos , Linfocitos Infiltrantes de Tumor , Inteligencia Artificial , Pronóstico
5.
Histopathology ; 85(3): 478-488, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39004603

RESUMEN

AIMS: Over 50% of breast cancer cases are "Human epidermal growth factor receptor 2 (HER2) low breast cancer (BC)", characterized by HER2 immunohistochemistry (IHC) scores of 1+ or 2+ alongside no amplification on fluorescence in situ hybridization (FISH) testing. The development of new anti-HER2 antibody-drug conjugates (ADCs) for treating HER2-low breast cancers illustrates the importance of accurately assessing HER2 status, particularly HER2-low breast cancer. In this study we evaluated the performance of a deep-learning (DL) model for the assessment of HER2, including an assessment of the causes of discordances of HER2-Null between a pathologist and the DL model. We specifically focussed on aligning the DL model rules with the ASCO/CAP guidelines, including stained cells' staining intensity and completeness of membrane staining. METHODS AND RESULTS: We trained a DL model on a multicentric cohort of breast cancer cases with HER2-IHC scores (n = 299). The model was validated on two independent multicentric validation cohorts (n = 369 and n = 92), with all cases reviewed by three senior breast pathologists. All cases underwent a thorough review by three senior breast pathologists, with the ground truth determined by a majority consensus on the final HER2 score among the pathologists. In total, 760 breast cancer cases were utilized throughout the training and validation phases of the study. The model's concordance with the ground truth (ICC = 0.77 [0.68-0.83]; Fisher P = 1.32e-10) is higher than the average agreement among the three senior pathologists (ICC = 0.45 [0.17-0.65]; Fisher P = 2e-3). In the two validation cohorts, the DL model identifies 95% [93% - 98%] and 97% [91% - 100%] of HER2-low and HER2-positive tumours, respectively. Discordant results were characterized by morphological features such as extended fibrosis, a high number of tumour-infiltrating lymphocytes, and necrosis, whilst some artefacts such as nonspecific background cytoplasmic stain in the cytoplasm of tumour cells also cause discrepancy. CONCLUSION: Deep learning can support pathologists' interpretation of difficult HER2-low cases. Morphological variables and some specific artefacts can cause discrepant HER2-scores between the pathologist and the DL model.


Asunto(s)
Biomarcadores de Tumor , Neoplasias de la Mama , Aprendizaje Profundo , Inmunohistoquímica , Receptor ErbB-2 , Humanos , Neoplasias de la Mama/patología , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/genética , Receptor ErbB-2/metabolismo , Receptor ErbB-2/genética , Femenino , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Patólogos , Hibridación Fluorescente in Situ , Persona de Mediana Edad
6.
JAMA ; 331(13): 1135-1144, 2024 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-38563834

RESUMEN

Importance: The association of tumor-infiltrating lymphocyte (TIL) abundance in breast cancer tissue with cancer recurrence and death in patients with early-stage triple-negative breast cancer (TNBC) who are not treated with adjuvant or neoadjuvant chemotherapy is unclear. Objective: To study the association of TIL abundance in breast cancer tissue with survival among patients with early-stage TNBC who were treated with locoregional therapy but no chemotherapy. Design, Setting, and Participants: Retrospective pooled analysis of individual patient-level data from 13 participating centers in North America (Rochester, Minnesota; Vancouver, British Columbia, Canada), Europe (Paris, Lyon, and Villejuif, France; Amsterdam and Rotterdam, the Netherlands; Milan, Padova, and Genova, Italy; Gothenburg, Sweden), and Asia (Tokyo, Japan; Seoul, Korea), including 1966 participants diagnosed with TNBC between 1979 and 2017 (with follow-up until September 27, 2021) who received treatment with surgery with or without radiotherapy but no adjuvant or neoadjuvant chemotherapy. Exposure: TIL abundance in breast tissue from resected primary tumors. Main Outcomes and Measures: The primary outcome was invasive disease-free survival [iDFS]. Secondary outcomes were recurrence-free survival [RFS], survival free of distant recurrence [distant RFS, DRFS], and overall survival. Associations were assessed using a multivariable Cox model stratified by participating center. Results: This study included 1966 patients with TNBC (median age, 56 years [IQR, 39-71]; 55% had stage I TNBC). The median TIL level was 15% (IQR, 5%-40%). Four-hundred seventeen (21%) had a TIL level of 50% or more (median age, 41 years [IQR, 36-63]), and 1300 (66%) had a TIL level of less than 30% (median age, 59 years [IQR, 41-72]). Five-year DRFS for stage I TNBC was 94% (95% CI, 91%-96%) for patients with a TIL level of 50% or more, compared with 78% (95% CI, 75%-80%) for those with a TIL level of less than 30%; 5-year overall survival was 95% (95% CI, 92%-97%) for patients with a TIL level of 50% or more, compared with 82% (95% CI, 79%-84%) for those with a TIL level of less than 30%. At a median follow-up of 18 years, and after adjusting for age, tumor size, nodal status, histological grade, and receipt of radiotherapy, each 10% higher TIL increment was associated independently with improved iDFS (hazard ratio [HR], 0.92 [0.89-0.94]), RFS (HR, 0.90 [0.87-0.92]), DRFS (HR, 0.87 [0.84-0.90]), and overall survival (0.88 [0.85-0.91]) (likelihood ratio test, P < 10e-6). Conclusions and Relevance: In patients with early-stage TNBC who did not undergo adjuvant or neoadjuvant chemotherapy, breast cancer tissue with a higher abundance of TIL levels was associated with significantly better survival. These results suggest that breast tissue TIL abundance is a prognostic factor for patients with early-stage TNBC.


Asunto(s)
Linfocitos Infiltrantes de Tumor , Neoplasias de la Mama Triple Negativas , Adulto , Humanos , Persona de Mediana Edad , Adyuvantes Inmunológicos , Colombia Británica , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/patología , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/patología , Estudios Retrospectivos , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/mortalidad , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/terapia
8.
NPJ Breast Cancer ; 10(1): 52, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38942745

RESUMEN

Tumor-Infiltrating Lymphocytes (TILs) have strong prognostic and predictive value in breast cancer, but their visual assessment is subjective. To improve reproducibility, the International Immuno-oncology Working Group recently released recommendations for the computational assessment of TILs that build on visual scoring guidelines. However, existing resources do not adequately address these recommendations due to the lack of annotation datasets that enable joint, panoptic segmentation of tissue regions and cells. Moreover, existing deep-learning methods focus entirely on either tissue segmentation or cell nuclei detection, which complicates the process of TILs assessment by necessitating the use of multiple models and reconciling inconsistent predictions. We introduce PanopTILs, a region and cell-level annotation dataset containing 814,886 nuclei from 151 patients, openly accessible at: sites.google.com/view/panoptils . Using PanopTILs we developed MuTILs, a neural network optimized for assessing TILs in accordance with clinical recommendations. MuTILs is a concept bottleneck model designed to be interpretable and to encourage sensible predictions at multiple resolutions. Using a rigorous internal-external cross-validation procedure, MuTILs achieves an AUROC of 0.93 for lymphocyte detection and a DICE coefficient of 0.81 for tumor-associated stroma segmentation. Our computational score closely matched visual scores from 2 pathologists (Spearman R = 0.58-0.61, p < 0.001). Moreover, computational TILs scores had a higher prognostic value than visual scores, independent of TNM stage and patient age. In conclusion, we introduce a comprehensive open data resource and a modeling approach for detailed mapping of the breast tumor microenvironment.

9.
Trends Cancer ; 10(6): 490-506, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38521654

RESUMEN

Triple-negative breast cancers (TNBCs) exhibit heightened T cell infiltration, contributing to an enhanced response to immune checkpoint blockade (ICB) compared with other subtypes. An immune-rich immune microenvironment correlates with improved prognosis in early and advanced TNBC. Combination chemotherapy and ICB is now the standard of care in early- and late-stage TNBC. Although programmed death ligand-1 (PD-L1) positivity predicts ICB response in advanced stages, its role in early-stage disease remains uncertain. Despite neoadjuvant ICB becoming common in early-stage TNBC, the necessity of adjuvant ICB after surgery remains unclear. Understanding the molecular basis of the immune response in breast cancer is vital for precise biomarkers for ICB and effective combination therapy strategies.


Asunto(s)
Biomarcadores de Tumor , Inhibidores de Puntos de Control Inmunológico , Terapia Neoadyuvante , Neoplasias de la Mama Triple Negativas , Microambiente Tumoral , Humanos , Femenino , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/terapia , Neoplasias de la Mama Triple Negativas/patología , Biomarcadores de Tumor/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de los fármacos , Terapia Neoadyuvante/métodos , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Antígeno B7-H1/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Pronóstico , Quimioterapia Adyuvante/métodos
10.
Nat Rev Cancer ; 24(8): 554-577, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38969810

RESUMEN

The tumour immune microenvironment is shaped by the crosstalk between cancer cells, immune cells, fibroblasts, endothelial cells and other stromal components. Although the immune tumour microenvironment (TME) serves as a source of therapeutic targets, it is also considered a friend or foe to tumour-directed therapies. This is readily illustrated by the importance of T cells in triple-negative breast cancer (TNBC), culminating in the advent of immune checkpoint therapy in combination with cytotoxic chemotherapy as standard of care for both early and advanced-stage TNBC, as well as recent promising signs of efficacy in a subset of hormone receptor-positive disease. In this Review, we discuss the various components of the immune TME in breast cancer and therapies that target or impact the immune TME, as well as the complexity of host physiology.


Asunto(s)
Microambiente Tumoral , Humanos , Microambiente Tumoral/inmunología , Femenino , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunoterapia/métodos
11.
NPJ Breast Cancer ; 10(1): 25, 2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38553444

RESUMEN

Operable triple-negative breast cancer (TNBC) has a higher risk of recurrence and death compared to other subtypes. Tumor size and nodal status are the primary clinical factors used to guide systemic treatment, while biomarkers of proliferation have not demonstrated value. Recent studies suggest that subsets of TNBC have a favorable prognosis, even without systemic therapy. We evaluated the association of fully automated mitotic spindle hotspot (AMSH) counts with recurrence-free (RFS) and overall survival (OS) in two separate cohorts of patients with early-stage TNBC who did not receive systemic therapy. AMSH counts were obtained from areas with the highest mitotic density in digitized whole slide images processed with a convolutional neural network trained to detect mitoses. In 140 patients from the Mayo Clinic TNBC cohort, AMSH counts were significantly associated with RFS and OS in a multivariable model controlling for nodal status, tumor size, and tumor-infiltrating lymphocytes (TILs) (p < 0.0001). For every 10-point increase in AMSH counts, there was a 16% increase in the risk of an RFS event (HR 1.16, 95% CI 1.08-1.25), and a 7% increase in the risk of death (HR 1.07, 95% CI 1.00-1.14). We corroborated these findings in a separate cohort of systemically untreated TNBC patients from Radboud UMC in the Netherlands. Our findings suggest that AMSH counts offer valuable prognostic information in patients with early-stage TNBC who did not receive systemic therapy, independent of tumor size, nodal status, and TILs. If further validated, AMSH counts could help inform future systemic therapy de-escalation strategies.

12.
NPJ Breast Cancer ; 10(1): 75, 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39169033

RESUMEN

Association of stromal tumor-infiltrating lymphocytes (sTILs) with survival outcomes among patients with metastatic breast cancer (MBC) remains unclear. The primary objective was to evaluate the association of sTILs with progression-free survival in randomized phase III trial CALGB 40502. sTILs were associated with progression-free and overall survival in chemotherapy-treated MBC when controlling for treatment arm; however, this effect did not remain significant after additional adjustment for hormone receptor status. CALGB is now part of the Alliance for Clinical Trials in Oncology. Trial Registration: ClinicalTrials.gov: NCT00785291.

13.
JAMA Oncol ; 10(8): 1077-1086, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38935352

RESUMEN

Importance: The absolute benefit of chemotherapy for all patients with stage I triple-negative breast cancer (TNBC) is unclear, and biomarkers are not currently available for selecting patients with an excellent outcome for whom neoadjuvant or adjuvant chemotherapy may have negligible benefit. High levels of stromal tumor-infiltrating lymphocytes (sTILs) are associated with favorable survival in TNBC, but data solely in stage I TNBC are lacking. Objective: To examine the outcomes of patients of all ages with stage I TNBC solely and who received neither neoadjuvant nor adjuvant chemotherapy, according to centrally reviewed sTIL levels at prespecified cutoffs. Design, Setting, and Participants: This cohort study used the Netherlands Cancer Registry to identify patients diagnosed with stage I TNBC between January 1, 2005, and December 31, 2015, who were not treated with chemotherapy. Only patients who did not receive neoadjuvant and/or adjuvant chemotherapy were selected. The clinical data were matched with their corresponding pathology data provided by the Dutch Pathology Registry. Data analysis was performed between February and October 2023. Main Outcomes and Measures: The primary end point was breast cancer-specific survival (BCSS) at 5, 10, and 15 years for the prespecified sTIL level cutoffs of 30%, 50%, and 75%. Hematoxylin and eosin-stained slides were used for central review of histologic subtype, grade, and lymphovascular invasion. The International Immuno-Oncology Biomarker Working Group guidelines were used to score the sTIL levels; these levels were determined for 1041 patients. Results: Of a total of 4511 females with stage I TNBC, patients who were not treated with chemotherapy were selected and tissue blocks requested; sTILs were scored in 1041 patients (mean [SD] age at diagnosis, 64.4 [11.1] years, median follow-up 11.4 [95% CI, 10.9-11.9] years) who were included in the analyses.. Most tumors (952 [91.5%]) were invasive carcinomas of nonspecial histologic subtype. Most patients (548 [52.6%]) had pT1cN0 tumors. Median (range) sTIL level was 5% (1%-99%). A total of 775 patients (74.4%) had sTIL levels below 30%, 266 (25.6%) had 30% or greater, 203 (19.5%) had 50% or greater, and 141 (13.5%) had 75% or greater. Patients with pT1abN0 tumors had a more favorable outcome vs patients with pT1cN0 tumors, with a 10-year BCSS of 92% (95% CI, 89%-94%) vs 86% (95% CI, 82%-89%). In the overall cohort, sTIL levels of at least 30% were associated with better BCSS compared with sTIL levels less than 30% (96% and 87%, respectively; hazard ratio [HR], 0.45; 95% CI, 0.26-0.77). High sTIL levels of 50% or greater were associated with a better outcome than low sTIL levels of less than 50% (HR, 0.27; 95% CI, 0.10-0.74) in patients with pT1C tumors, with a 10-year BCSS of 95% increasing to 98% with sTIL levels of 75% or greater. Conclusions and Relevance: Results of this study showed that patients with stage I TNBC and high level of sTILs who did not receive neoadjuvant or adjuvant chemotherapy had excellent 10-year BCSS. The findings further support the role of sTILs as integral biomarkers in prospective clinical trials of therapy optimization for this patient population.


Asunto(s)
Linfocitos Infiltrantes de Tumor , Estadificación de Neoplasias , Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/mortalidad , Linfocitos Infiltrantes de Tumor/inmunología , Femenino , Persona de Mediana Edad , Anciano , Adulto , Países Bajos , Terapia Neoadyuvante , Quimioterapia Adyuvante , Estudios de Cohortes
14.
Eur J Cancer ; 209: 114225, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39053288

RESUMEN

Precision oncology has a significant role to play in delivering optimal patient care. Biomarkers are critical enablers for precision oncology across the continuum of cancer diagnosis, in defining patient prognosis, and in predicting the response to treatments and their potential toxicities, as well as delineating the risk of hereditary cancer syndromes. Biomarkers also potentiate cancer drug development, accelerating patient access to safe and effective therapies. However, despite an accurate and timely diagnosis being critical to patient survival, advances in genomic testing are not being fully exploited in daily clinical practice, leading to missed opportunities to deliver the most effective treatments for patients. Biomarker testing availability and implementation often lag behind approvals of respective biomarker-informed therapies, limiting prompt patient access to these life-saving drugs. Multiple factors currently impede the routine adoption of biomarker testing including, but not limited to, cost, lack of test reimbursement, limited access, regulatory hurdles, lack of knowledge, insufficient cooperation on assay development, and the urgent need to harmonize and validate testing assays, all leading to inefficient diagnostic pathways. Clinical guidelines increasingly include genomic profiling, and recent evidence suggests that precision oncology can be delivered in a cost-effective way for financially-challenged health systems. Therefore, precision genomic testing for cancer biomarkers must be embedded into the clinical practice of oncology care delivery going forward. We articulate a series of recommendations and a call to action to underpin the mainstreaming of a biomarker-informed precision oncology approach to enhance patient outcomes and deliver cost effective 21st century cancer care.

15.
Clin Cancer Res ; 30(10): 2160-2169, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38466643

RESUMEN

PURPOSE: Stromal tumor-infiltrating lymphocytes (sTIL) are associated with pathologic complete response (pCR) and long-term outcomes for triple-negative breast cancer (TNBC) in the setting of anthracycline-based chemotherapy. The impact of sTILs on refining outcomes beyond prognostic information provided by pCR in anthracycline-free neoadjuvant chemotherapy (NAC) is not known. EXPERIMENTAL DESIGN: This is a pooled analysis of two studies where patients with stage I (T>1 cm)-III TNBC received carboplatin (AUC 6) plus docetaxel (75 mg/m2; CbD) NAC. sTILs were evaluated centrally on pre-treatment hematoxylin and eosin slides using standard criteria. Cox regression analysis was used to examine the effect of variables on event-free survival (EFS) and overall survival (OS). RESULTS: Among 474 patients, 44% had node-positive disease. Median sTILs were 5% (range, 1%-95%), and 32% of patients had ≥30% sTILs. pCR rate was 51%. On multivariable analysis, T stage (OR, 2.08; P = 0.007), nodal status (OR, 1.64; P = 0.035), and sTILs (OR, 1.10; P = 0.011) were associated with pCR. On multivariate analysis, nodal status (HR, 0.46; P = 0.008), pCR (HR, 0.20; P < 0.001), and sTILs (HR, 0.95; P = 0.049) were associated with OS. At 30% cut-point, sTILs stratified outcomes in stage III disease, with 5-year OS 86% versus 57% in ≥30% versus <30% sTILs (HR, 0.29; P = 0.014), and numeric trend in stage II, with 5-year OS 93% versus 89% in ≥30% versus <30% sTILs (HR, 0.55; P = 0.179). Among stage II-III patients with pCR, EFS was better in those with ≥30% sTILs (HR, 0.16; P, 0.047). CONCLUSIONS: sTILs density was an independent predictor of OS beyond clinicopathologic features and pathologic response in patients with TNBC treated with anthracycline-free CbD chemotherapy. Notably, sTILs density stratified outcomes beyond tumor-node-metastasis (TNM) stage and pathologic response. These findings highlight the role of sTILs in patient selection and stratification for neo/adjuvant escalation and de-escalation strategies.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Linfocitos Infiltrantes de Tumor , Terapia Neoadyuvante , Neoplasias de la Mama Triple Negativas , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Femenino , Terapia Neoadyuvante/métodos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/mortalidad , Neoplasias de la Mama Triple Negativas/patología , Persona de Mediana Edad , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Adulto , Anciano , Antraciclinas/administración & dosificación , Antraciclinas/uso terapéutico , Pronóstico , Estadificación de Neoplasias , Resultado del Tratamiento , Docetaxel/administración & dosificación , Docetaxel/uso terapéutico , Carboplatino/administración & dosificación
16.
Cancer Discov ; 14(6): 1018-1047, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38581685

RESUMEN

Understanding the role of the tumor microenvironment (TME) in lung cancer is critical to improving patient outcomes. We identified four histology-independent archetype TMEs in treatment-naïve early-stage lung cancer using imaging mass cytometry in the TRACERx study (n = 81 patients/198 samples/2.3 million cells). In immune-hot adenocarcinomas, spatial niches of T cells and macrophages increased with clonal neoantigen burden, whereas such an increase was observed for niches of plasma and B cells in immune-excluded squamous cell carcinomas (LUSC). Immune-low TMEs were associated with fibroblast barriers to immune infiltration. The fourth archetype, characterized by sparse lymphocytes and high tumor-associated neutrophil (TAN) infiltration, had tumor cells spatially separated from vasculature and exhibited low spatial intratumor heterogeneity. TAN-high LUSC had frequent PIK3CA mutations. TAN-high tumors harbored recently expanded and metastasis-seeding subclones and had a shorter disease-free survival independent of stage. These findings delineate genomic, immune, and physical barriers to immune surveillance and implicate neutrophil-rich TMEs in metastasis. SIGNIFICANCE: This study provides novel insights into the spatial organization of the lung cancer TME in the context of tumor immunogenicity, tumor heterogeneity, and cancer evolution. Pairing the tumor evolutionary history with the spatially resolved TME suggests mechanistic hypotheses for tumor progression and metastasis with implications for patient outcome and treatment. This article is featured in Selected Articles from This Issue, p. 897.


Asunto(s)
Neoplasias Pulmonares , Microambiente Tumoral , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/genética , Microambiente Tumoral/inmunología , Linfocitos T/inmunología , Células Mieloides/inmunología , Femenino , Masculino , Evasión Inmune
18.
Rev. argent. cir ; 112(4): 450-458, dic. 2020. graf, tab
Artículo en Español | LILACS, BINACIS | ID: biblio-1288157

RESUMEN

RESUMEN Antecedentes: la pandemia por COVID-19 generó cambios en el manejo de pacientes con todo tipo de patologías. Mostramos la experiencia respecto de los pacientes con apendicitis aguda (AA) operados en un hospital universitario de la Ciudad Autónoma de Buenos Aires durante los meses de confina miento y su comparación con el mismo período del año 2019 a fin de evaluar las diferencias. Material y métodos: se analizó en forma retrospectiva una base confeccionada de forma prospectiva. Se compararon pacientes operados de AA, consignando los datos respecto del período preoperatorio, comparando datos inherentes a la cirugía y sus resultados en los períodos marzo-julio de 2019 y 2020. Resultados: fueron incluidos 127 pacientes, 46 de ellos operados durante la pandemia. Los pacientes operados durante la pandemia presentaron mayor incidencia de peritonitis (61,7% vs. 76,1%; p: 0,09) y mayor requerimiento de drenaje abdominal (9,9% vs. 23,9%; p: 0,03). También tuvieron mayor in cidencia de complicaciones (10,9 vs. 4,9%; p: 0,21), reoperaciones (8,7 vs. 1,23%, p: 0,03), reinterna ciones (6,5 vs. 0%, p: 0,02) y requerimiento de antibioticoterapia endovenosa prolongada (6,5 vs. 0%; p: 0,02). La estadía hospitalaria fue mayor para la cohorte operada durante la pandemia: 3,24 días (Desvío estándar [D.E.]: 7,31) vs. 1,89 días (D.E: 2,04). Conclusiones: durante la pandemia por COVID-19, el número de pacientes operados por AA disminuyó con respecto al año 2019; se observaron estadios más avanzados de la enfermedad, mayores compli caciones en el posoperatorio y mayor estadía hospitalaria.


ABSTRACT Background: COVID-19 pandemic has generated changes regarding the management of patients with all kind of pathologies. Here we show the experience concerning the surgical treatment of acute appendicitis (AA) in a teaching hospital in the city of Buenos Aires during the lockdown, and its comparison with the same period of 2019 in order to assess the differences between the timing for consultation, intraoperative findings and the treatment outcome. Material and methods: A prospective database was retrospectively analyzed. We compared patients undergoing surgical treatment for AA in March-July of 2019 v. March-July 2020, taking into account the preoperative period and comparing the outcome regarding the surgical intervention and the impact in the postoperative period between the two groups. Results: 127 patients were included, 46 treated during the pandemic. Patients undergoing surgical treatment during the pandemic had a higher incidence of peritonitis (61.7% vs. 76.1%; p = 0.09) and higher requirement of abdominal drainage (9.9% vs. 23.9%; p = 0.03). There was an increase in the incidence of complications (10.9 vs. 4.9%; p: 0.21), reoperations (8.7 vs. 1.23%, p: 0.03), readmissions (6.5 vs. 0%, p: 0.02) and need for out-patient intravenous antibiotics (6.5 vs. 0%; p: 0.02). The average hospital length was of 1.89 (SD 2.04) and 3.24 (SD 7.31) for the groups operated before and during pandemic, respectively. Conclusions: During the COVID-19 pandemic, the number of patients undergoing surgical intervention because of AA decreased compared to the previous year, with increased complications in the postoperative period, higher compromise of the appendix, and longer hospital stay.


Asunto(s)
Humanos , Masculino , Femenino , Adulto , Persona de Mediana Edad , Adulto Joven , Apendicectomía/estadística & datos numéricos , Apendicitis/cirugía , COVID-19 , Apendicitis/complicaciones , Peritonitis , Servicios Médicos de Urgencia/estadística & datos numéricos , Pandemias
20.
Salud(i)ciencia (Impresa) ; 13(1): 17-20, 2004. graf.
Artículo en Español | LILACS | ID: biblio-1359381

RESUMEN

Thromboembolic complications are often associated with high morbidity and mortality in patients with cancer. Considerable evidence exists that intra-tumoural activation of coagulation provides growth-promoting properties to the neoplasm. Fibrinolysis is an active part of stromal remodelling. Fibrin is degraded by tumour-derived plasmin in fragments, of which Ddimers recently spurred most interest. We analysed in 96 patients with breast cancer the levels of circulating D-dimers. D-dimer levels were positively correlated with tumour load, number of metastatic sites, progression kinetics and the cytokines related to angiogenesis: serum VEGF, calculated VEGF load in platelets and serum interleukin-6 (IL-6). In a further study we measured the circulating angiogenic factors VEGF-A, IL-6 and the fibrin D-dimer fragment in the draining veins of tumours in 21 cancer patients. Our data suggest that IL-6, but not serum VEGF is tumour-derived. In the megakaryoblastic cell line MEG-01, the expression of VEGFA was regulated by IL-6. Thus, the higher platelet VEGF-A load resulting in higher serum VEGF levels in cancer patients may partly result from an IL-6 mediated upregulation of the expression of VEGF-A in the precursor of the platelet, i.e. the megakaryocyte. We further confirmed that platelets adhere and aggregate on tumour endothelium. We propose that IL-6 indirectly promotes tumour angiogenesis through its upregulation of the VEGF-A load in platelets. In addition, the correlations found between peripheral venous IL-6 and peripheral venous fibrinogen and D-dimers levels, and the high D-dimer levels found in the draining vein of the tumours suggest an important contribution for IL-6 in extravascular fibrinogen metabolism. Our results suggest a pivotal role for IL-6 in the intrinsic link between haemostasis and angiogenesis.


Las complicaciones tromboembólicas a menudo se asocian con mayor mortalidad y morbilidad en pacientes con cáncer. Existen indicios de que la activación intratumoral de la coagulación tiene propiedades que promueven el crecimiento de la neoplasia. La fibrinólisis es una parte importante del remodelamiento del estroma. La fibrina es degradada por la plasmina derivada del tumor en fragmentos, entre los cuales los D-dímeros motivan el mayor interés. Estudiamos en 96 pacientes con cáncer de mama los niveles circulantes de D-dímeros. Su concentración se correlacionó en forma positiva con la carga tumoral, el número de sitios metastásicos, la cinética de progresión y las citoquinas liberadas durante la angiogénesis: VEGF sérico, carga calculada de VEGF en plaquetas y concentración sérica de interleuquina (IL) 6. En un estudio posterior medimos los factores angiogénicos circulantes, VEGF-A, IL-6 y el fragmento D-dímero de fibrina en el drenaje venoso de tumores en 21 pacientes con cáncer. Nuestros resultados sugieren que la IL-6, pero no el VEGF, deriva del tumor. En la línea celular megacarioblástica MEG-01, la expresión de VEGF-A estuvo regulada por la IL-6. Por ende, la mayor carga plaquetaria de VEGF-A que se asocia con mayor concentración sérica de VEGF en pacientes con cáncer puede ser en parte consecuencia de la mayor expresión inducida por IL-6 en los precursores plaquetarios, como megacariocitos. Posteriormente confirmamos que las plaquetas se agregan y se adhieren al endotelio tumoral. Proponemos que la IL-6 promueve en forma indirecta la angiogénesis del tumor a través de la estimulación de la producción de VEGF-A en plaquetas. Además, la correlación encontrada entre la IL-6, fibrinógeno y D-dímeros en sangre venosa periférica y la concentración elevada de D-dímero en venas de drenaje tumoral sugiere una importante contribución de la IL-6 en el metabolismo extravascular del fibrinógeno. Nuestras observaciones sugieren un papel crucial de la IL-6 en la conexión intrínseca entre hemostasia y angiogénesis.


Asunto(s)
Interleucina-6 , Hemostasis , Sangre , Coagulación Sanguínea , Neoplasias de la Mama , Morbilidad , Factor A de Crecimiento Endotelial Vascular , Neoplasias
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA