Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 615(7950): 134-142, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36470304

RESUMEN

Preventing SARS-CoV-2 infection by modulating viral host receptors, such as angiotensin-converting enzyme 2 (ACE2)1, could represent a new chemoprophylactic approach for COVID-19 that complements vaccination2,3. However, the mechanisms that control the expression of ACE2 remain unclear. Here we show that the farnesoid X receptor (FXR) is a direct regulator of ACE2 transcription in several tissues affected by COVID-19, including the gastrointestinal and respiratory systems. We then use the over-the-counter compound z-guggulsterone and the off-patent drug ursodeoxycholic acid (UDCA) to reduce FXR signalling and downregulate ACE2 in human lung, cholangiocyte and intestinal organoids and in the corresponding tissues in mice and hamsters. We show that the UDCA-mediated downregulation of ACE2 reduces susceptibility to SARS-CoV-2 infection in vitro, in vivo and in human lungs and livers perfused ex situ. Furthermore, we reveal that UDCA reduces the expression of ACE2 in the nasal epithelium in humans. Finally, we identify a correlation between UDCA treatment and positive clinical outcomes after SARS-CoV-2 infection using retrospective registry data, and confirm these findings in an independent validation cohort of recipients of liver transplants. In conclusion, we show that FXR has a role in controlling ACE2 expression and provide evidence that modulation of this pathway could be beneficial for reducing SARS-CoV-2 infection, paving the way for future clinical trials.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , COVID-19 , Receptores Virales , Ácido Ursodesoxicólico , Animales , Humanos , Ratones , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/metabolismo , COVID-19/prevención & control , Receptores Virales/genética , Receptores Virales/metabolismo , Estudios Retrospectivos , SARS-CoV-2/metabolismo , Tratamiento Farmacológico de COVID-19 , Cricetinae , Transcripción Genética , Ácido Ursodesoxicólico/farmacología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Organoides/efectos de los fármacos , Organoides/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/metabolismo , Sistema de Registros , Reproducibilidad de los Resultados , Trasplante de Hígado
2.
Liver Transpl ; 21(3): 314-20, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25488693

RESUMEN

Atrial fibrillation (AF) is the most common cardiac arrhythmia, and it is associated with increased cardiovascular morbidity and all-cause mortality. Our aim was to determine the impact of preexisting AF on patients undergoing liver transplantation (LT). A retrospective case-control study was performed. Records from patients who underwent LT between January 2005 and December 2008 at Mayo Clinic Florida were reviewed. Patients with preexisting AF were identified and matched to patients who did not have a diagnosis of AF. Thirty-two of 717 LT recipients (4.5%) had AF before LT. These patients were compared to a control group of 63 LT recipients. Pre-LT left ventricular hypertrophy (P = 0.03), a history of congestive heart failure (P = 0.04), and a history of stroke or transient ischemic attack (P = 0.03) were significantly more prevalent in patients with AF versus controls. Intraoperative adverse cardiac events (P = 0.02) and AF-related adverse postoperative events (P < 0.001) were more common in the recipients with known AF. Six patients with paroxysmal AF (19%) developed chronic/persistent AF postoperatively. Graft survival and patient survival were similar in the groups. Although patients with AF had a higher incidence of intraoperative cardiac events, a higher cardiovascular morbidity rate, and a complicated postoperative course, this did not affect overall graft and patient survival.


Asunto(s)
Fibrilación Atrial/complicaciones , Enfermedad Hepática en Estado Terminal/cirugía , Cardiopatías/etiología , Trasplante de Hígado/efectos adversos , Adulto , Anciano , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/mortalidad , Enfermedad Hepática en Estado Terminal/complicaciones , Enfermedad Hepática en Estado Terminal/diagnóstico , Enfermedad Hepática en Estado Terminal/mortalidad , Femenino , Florida , Supervivencia de Injerto , Cardiopatías/diagnóstico , Cardiopatías/mortalidad , Humanos , Incidencia , Estimación de Kaplan-Meier , Trasplante de Hígado/mortalidad , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento
3.
Liver Int ; 34(6): e105-10, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24529030

RESUMEN

BACKGROUND & AIMS: Non-ischaemic cardiomyopathy (NIC) is an early complication of liver transplantation (LT). Our aims were to define the prevalence, associated clinical factors, and prognosis of this condition. METHODS: A retrospective study was performed on patients undergoing LT at our institution from January 2005 to December 2012. Patients who developed NIC were identified. Data collected included demographic and clinical data. RESULTS: A total 1460 transplants were performed in this period and seventeen patients developed NIC. Pretransplant median QTc interval was 459 (range, 405-530), and median E/A ratio was 1 (range, 0.71-1.67). Fourteen patients (82%) were severely malnourished and required nutritional support. Thirteen patients (76%) had renal insufficiency. Median time to onset was 2 days post-transplant (range, 0-20). Echocardiograms showed global left ventricular hypokinesis and a decrease in ejection fraction (EF) from a median of 65% (range, 50-81) pretransplant to a median of 21% (range, 15-32). Median raw model for end-stage liver disease (MELD) score was 29 in patients with NIC vs. 18 in patients without cardiomyopathy (P = 0.01). There was no significant difference between recipients with NIC vs. recipients without cardiomyopathy regarding donor age, donor risk index, and cold and warm ischaemia time. Recovery of cardiac function occurred in 16 patients, with a median EF of 44% (range, 25-65%) at the time of discharge. The last echocardiogram available showed a median EF of 59% (range, 49-73%). One-year survival of NIC patients was 94.1%. CONCLUSION: Non-ischaemic cardiomyopathy is a rare complication after LT. Patients with NIC are critically ill, with high MELD score, and severe malnutrition.


Asunto(s)
Cardiomiopatías/etiología , Hepatopatías/cirugía , Trasplante de Hígado/efectos adversos , Disfunción Ventricular Izquierda/etiología , Función Ventricular Izquierda , Anciano , Cardiomiopatías/diagnóstico , Enfermedad Crítica , Femenino , Florida , Humanos , Hepatopatías/complicaciones , Hepatopatías/diagnóstico , Masculino , Desnutrición/complicaciones , Desnutrición/diagnóstico , Persona de Mediana Edad , Estado Nutricional , Recuperación de la Función , Estudios Retrospectivos , Factores de Riesgo , Índice de Severidad de la Enfermedad , Volumen Sistólico , Factores de Tiempo , Resultado del Tratamiento , Disfunción Ventricular Izquierda/diagnóstico , Disfunción Ventricular Izquierda/fisiopatología
4.
Cardiovasc Res ; 120(2): 174-187, 2024 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-38041432

RESUMEN

AIMS: Cardiac involvement is common in patients hospitalized with COVID-19 and correlates with an adverse disease trajectory. While cardiac injury has been attributed to direct viral cytotoxicity, serum-induced cardiotoxicity secondary to serological hyperinflammation constitutes a potentially amenable mechanism that remains largely unexplored. METHODS AND RESULTS: To investigate serological drivers of cardiotoxicity in COVID-19 we have established a robust bioassay that assessed the effects of serum from COVID-19 confirmed patients on human embryonic stem cell (hESC)-derived cardiomyocytes. We demonstrate that serum from COVID-19 positive patients significantly reduced cardiomyocyte viability independent of viral transduction, an effect that was also seen in non-COVID-19 acute respiratory distress syndrome (ARDS). Serum from patients with greater disease severity led to worse cardiomyocyte viability and this significantly correlated with levels of key inflammatory cytokines, including IL-6, TNF-α, IL1-ß, IL-10, CRP, and neutrophil to lymphocyte ratio with a specific reduction of CD4+ and CD8+ cells. Combinatorial blockade of IL-6 and TNF-α partly rescued the phenotype and preserved cardiomyocyte viability and function. Bulk RNA sequencing of serum-treated cardiomyocytes elucidated specific pathways involved in the COVID-19 response impacting cardiomyocyte viability, structure, and function. The observed effects of serum-induced cytotoxicity were cell-type selective as serum exposure did not adversely affect microvascular endothelial cell viability but resulted in endothelial activation and a procoagulant state. CONCLUSION: These results provide direct evidence that inflammatory cytokines are at least in part responsible for the cardiovascular damage seen in COVID-19 and characterise the downstream activated pathways in human cardiomyocytes. The serum signature of patients with severe disease indicates possible targets for therapeutic intervention.


Asunto(s)
COVID-19 , Humanos , Citocinas , Cardiotoxicidad , Interleucina-6 , Factor de Necrosis Tumoral alfa
5.
Stem Cell Reports ; 18(4): 936-951, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-37001515

RESUMEN

Ischemic heart failure is due to irreversible loss of cardiomyocytes. Preclinical studies showed that human pluripotent stem cell (hPSC)-derived cardiomyocytes could remuscularize infarcted hearts and improve cardiac function. However, these cardiomyocytes remained immature. Incorporating hPSC-derived epicardial cells has been shown to improve cardiomyocyte maturation, but the exact mechanisms are unknown. We posited epicardial fibronectin (FN1) as a mediator of epicardial-cardiomyocyte crosstalk and assessed its role in driving hPSC-derived cardiomyocyte maturation in 3D-engineered heart tissues (3D-EHTs). We found that the loss of FN1 with peptide inhibition F(pUR4), CRISPR-Cas9-mediated FN1 knockout, or tetracycline-inducible FN1 knockdown in 3D-EHTs resulted in immature cardiomyocytes with decreased contractile function, and inefficient Ca2+ handling. Conversely, when we supplemented 3D-EHTs with recombinant human FN1, we could recover hPSC-derived cardiomyocyte maturation. Finally, our RNA-sequencing analyses found FN1 within a wider paracrine network of epicardial-cardiomyocyte crosstalk, thus solidifying FN1 as a key driver of hPSC-derived cardiomyocyte maturation in 3D-EHTs.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Humanos , Miocitos Cardíacos , Fibronectinas , Diferenciación Celular/genética
6.
ESC Heart Fail ; 8(5): 4119-4129, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34390216

RESUMEN

AIMS: Membrane-bound angiotensin-converting enzyme (ACE)2 is the main cellular access point for SARS-CoV-2, but its expression and the effect of ACE inhibition have not been assessed quantitatively in patients with heart failure. The aim of this study was to characterize membrane-bound ACE2 expression in the myocardium and myocardial vasculature in patients undergoing heart transplantation and to assess the effect of pharmacological ACE inhibition. METHODS AND RESULTS: Left ventricular (LV) tissue was obtained from 36 explanted human hearts from patients undergoing heart transplantation. Immunohistochemical staining with antibodies directed against ACE2 co-registered with cardiac troponin T (cTnT) and α-smooth muscle cell actin (SMA) was performed across the entire cohort. ACE2 receptor expression was quantitatively assessed throughout the myocardium and vasculature. ACE2 was consistently expressed throughout the LV myocardium (28.3% ± 22.2% of cardiomyocytes). ACE2 expression was also detected in small calibre blood vessels (range, 2-9 µm), albeit at quantitatively much lower levels (5% ± 9% of blood vessels). There was no significant difference in ACE2 expression between patients receiving ACE inhibitors prior to transplantation and ACE inhibitor-negative controls (P > 0.05). ACE2 expression did not differ significantly between the different diagnostic groups as the underlying reason for heart transplantation (ANOVA > 0.05). N-terminal pro-brain natriuretic peptide (NT-proBNP) (R2  = 0.37, P = 0.0006) and pulmonary capillary wedge pressure (PCWP) (R2  = 0.13, P = 0.043) assessed by right heart catheterization were significantly correlated with greater ACE2 expression in cardiomyocytes. CONCLUSIONS: These data provide a comprehensive characterization of membrane-bound cardiac ACE2 expression in patients with heart failure with no demonstrable effect exerted by ACE inhibitors.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Trasplante de Corazón , Inhibidores de la Enzima Convertidora de Angiotensina , Humanos
8.
Stem Cell Reports ; 16(3): 478-492, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33657418

RESUMEN

COVID-19 patients often develop severe cardiovascular complications, but it remains unclear if these are caused directly by viral infection or are secondary to a systemic response. Here, we examine the cardiac tropism of SARS-CoV-2 in human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) and smooth muscle cells (hPSC-SMCs). We find that that SARS-CoV-2 selectively infects hPSC-CMs through the viral receptor ACE2, whereas in hPSC-SMCs there is minimal viral entry or replication. After entry into cardiomyocytes, SARS-CoV-2 is assembled in lysosome-like vesicles and egresses via bulk exocytosis. The viral transcripts become a large fraction of cellular mRNA while host gene expression shifts from oxidative to glycolytic metabolism and upregulates chromatin modification and RNA splicing pathways. Most importantly, viral infection of hPSC-CMs progressively impairs both their electrophysiological and contractile function, and causes widespread cell death. These data support the hypothesis that COVID-19-related cardiac symptoms can result from a direct cardiotoxic effect of SARS-CoV-2.


Asunto(s)
COVID-19/virología , Células Madre Pluripotentes Inducidas/virología , Miocitos Cardíacos/virología , SARS-CoV-2/patogenicidad , Células Cultivadas , Humanos , Empalme del ARN/genética , ARN Mensajero/genética , SARS-CoV-2/genética , Internalización del Virus
9.
Science ; 371(6531): 839-846, 2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-33602855

RESUMEN

Organoid technology holds great promise for regenerative medicine but has not yet been applied to humans. We address this challenge using cholangiocyte organoids in the context of cholangiopathies, which represent a key reason for liver transplantation. Using single-cell RNA sequencing, we show that primary human cholangiocytes display transcriptional diversity that is lost in organoid culture. However, cholangiocyte organoids remain plastic and resume their in vivo signatures when transplanted back in the biliary tree. We then utilize a model of cell engraftment in human livers undergoing ex vivo normothermic perfusion to demonstrate that this property allows extrahepatic organoids to repair human intrahepatic ducts after transplantation. Our results provide proof of principle that cholangiocyte organoids can be used to repair human biliary epithelium.


Asunto(s)
Enfermedades de los Conductos Biliares/terapia , Conductos Biliares Intrahepáticos/fisiología , Conductos Biliares/citología , Tratamiento Basado en Trasplante de Células y Tejidos , Células Epiteliales/citología , Organoides/trasplante , Animales , Bilis , Conductos Biliares/fisiología , Conductos Biliares Intrahepáticos/citología , Conducto Colédoco/citología , Células Epiteliales/fisiología , Vesícula Biliar/citología , Regulación de la Expresión Génica , Humanos , Hígado/fisiología , Trasplante de Hígado , Trasplante de Células Madre Mesenquimatosas , Ratones , Organoides/fisiología , RNA-Seq , Obtención de Tejidos y Órganos , Transcriptoma
10.
Nat Biotechnol ; 37(8): 895-906, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31375810

RESUMEN

The epicardium and its derivatives provide trophic and structural support for the developing and adult heart. Here we tested the ability of human embryonic stem cell (hESC)-derived epicardium to augment the structure and function of engineered heart tissue in vitro and to improve efficacy of hESC-cardiomyocyte grafts in infarcted athymic rat hearts. Epicardial cells markedly enhanced the contractility, myofibril structure and calcium handling of human engineered heart tissues, while reducing passive stiffness compared with mesenchymal stromal cells. Transplanted epicardial cells formed persistent fibroblast grafts in infarcted hearts. Cotransplantation of hESC-derived epicardial cells and cardiomyocytes doubled graft cardiomyocyte proliferation rates in vivo, resulting in 2.6-fold greater cardiac graft size and simultaneously augmenting graft and host vascularization. Notably, cotransplantation improved systolic function compared with hearts receiving either cardiomyocytes alone, epicardial cells alone or vehicle. The ability of epicardial cells to enhance cardiac graft size and function makes them a promising adjuvant therapeutic for cardiac repair.


Asunto(s)
Corazón/fisiología , Células Madre Embrionarias Humanas , Infarto del Miocardio/terapia , Miocitos Cardíacos , Regeneración , Animales , Embrión de Pollo , Regulación de la Expresión Génica , Humanos , Masculino , Ratas , Ratas Desnudas , Ratas Sprague-Dawley , Ingeniería de Tejidos
11.
Am J Cardiol ; 119(5): 687-691, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27865482

RESUMEN

Cardiac rehabilitation (CR) improves exercise capacity (EC), but not all CR participants achieve such improvements. Our primary aim was to develop a tool to identify those with suboptimal improvement in EC after CR. We retrospectively analyzed 541 patients enrolled in a phase-II CR program after a cardiac event or intervention from 2003 to 2014. EC was assessed with the 6-minute walk test. We developed a multivariate linear regression model and corresponding nomogram to predict EC after CR. The predictors included in the final model were age, gender, baseline EC, primary referral diagnosis, body mass index, systolic blood pressure at rest, triglycerides, low-density lipoprotein cholesterol, lipid-lowering medication use, and an interaction term of low-density lipoprotein cholesterol with lipid-lowering therapy. The prediction model was internally validated using bootstrap methods, and a nomogram was created for ease of use. In conclusion, this tool helps to identify those patients with suboptimal improvement in EC who could be targeted for individualized interventions to increase their performance.


Asunto(s)
Angina Estable/rehabilitación , Rehabilitación Cardiaca , Puente de Arteria Coronaria/rehabilitación , Tolerancia al Ejercicio , Infarto del Miocardio/rehabilitación , Intervención Coronaria Percutánea/rehabilitación , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Presión Sanguínea , Índice de Masa Corporal , Procedimientos Quirúrgicos Cardíacos/rehabilitación , LDL-Colesterol/sangre , Femenino , Válvulas Cardíacas/cirugía , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Hipolipemiantes/uso terapéutico , Modelos Lineales , Masculino , Persona de Mediana Edad , Análisis Multivariante , Nomogramas , Pronóstico , Reproducibilidad de los Resultados , Estudios Retrospectivos , Factores Sexuales , Resultado del Tratamiento , Triglicéridos/sangre , Prueba de Paso
12.
Nat Med ; 23(8): 954-963, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28671689

RESUMEN

The treatment of common bile duct (CBD) disorders, such as biliary atresia or ischemic strictures, is restricted by the lack of biliary tissue from healthy donors suitable for surgical reconstruction. Here we report a new method for the isolation and propagation of human cholangiocytes from the extrahepatic biliary tree in the form of extrahepatic cholangiocyte organoids (ECOs) for regenerative medicine applications. The resulting ECOs closely resemble primary cholangiocytes in terms of their transcriptomic profile and functional properties. We explore the regenerative potential of these organoids in vivo and demonstrate that ECOs self-organize into bile duct-like tubes expressing biliary markers following transplantation under the kidney capsule of immunocompromised mice. In addition, when seeded on biodegradable scaffolds, ECOs form tissue-like structures retaining biliary characteristics. The resulting bioengineered tissue can reconstruct the gallbladder wall and repair the biliary epithelium following transplantation into a mouse model of injury. Furthermore, bioengineered artificial ducts can replace the native CBD, with no evidence of cholestasis or occlusion of the lumen. In conclusion, ECOs can successfully reconstruct the biliary tree, providing proof of principle for organ regeneration using human primary cholangiocytes expanded in vitro.


Asunto(s)
Conductos Biliares Extrahepáticos/fisiología , Células Epiteliales/citología , Vesícula Biliar/fisiología , Organoides/fisiología , Regeneración/fisiología , Ingeniería de Tejidos/métodos , Animales , Conductos Biliares Extrahepáticos/citología , Conductos Biliares Extrahepáticos/lesiones , Sistema Biliar/citología , Sistema Biliar/lesiones , Sistema Biliar/fisiología , Trasplante de Células , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Vesícula Biliar/lesiones , Humanos , Técnicas In Vitro , Queratina-19/metabolismo , Queratina-7/metabolismo , Ratones , Organoides/citología , Organoides/efectos de los fármacos , Organoides/metabolismo , Secretina/farmacología , Somatostatina/farmacología , Andamios del Tejido , gamma-Glutamiltransferasa/metabolismo
13.
Stem Cells Transl Med ; 5(7): 946-59, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27194743

RESUMEN

UNLABELLED: Vascular smooth muscle cells (SMCs) from distinct anatomic locations derive from different embryonic origins. Here we investigated the respective potential of different embryonic origin-specific SMCs derived from human embryonic stem cells (hESCs) to support endothelial network formation in vitro. SMCs of three distinct embryological origins were derived from an mStrawberry-expressing hESC line and were cocultured with green fluorescent protein-expressing human umbilical vein endothelial cells (HUVECs) to investigate the effects of distinct SMC subtypes on endothelial network formation. Quantitative analysis demonstrated that lateral mesoderm (LM)-derived SMCs best supported HUVEC network complexity and survival in three-dimensional coculture in Matrigel. The effects of the LM-derived SMCs on HUVECs were at least in part paracrine in nature. A TaqMan array was performed to identify the possible mediators responsible for the differential effects of the SMC lineages, and a microarray was used to determine lineage-specific angiogenesis gene signatures. Midkine (MDK) was identified as one important mediator for the enhanced vasculogenic potency of LM-derived SMCs. The functional effects of MDK on endothelial network formation were then determined by small interfering RNA-mediated knockdown in SMCs, which resulted in impaired network complexity and survival of LM-derived SMC cocultures. The present study is the first to show that SMCs from distinct embryonic origins differ in their ability to support HUVEC network formation. LM-derived SMCs best supported endothelial cell network complexity and survival in vitro, in part through increased expression of MDK. A lineage-specific approach might be beneficial for vascular tissue engineering and therapeutic revascularization. SIGNIFICANCE: Mural cells are essential for the stabilization and maturation of new endothelial cell networks. However, relatively little is known of the effect of the developmental origins of mural cells on their signaling to endothelial cells and how this affects vessel development. The present study demonstrated that human smooth muscle cells (SMCs) from distinct embryonic origins differ in their ability to support endothelial network formation. Lateral mesoderm-derived SMCs best support endothelial cell network complexity and survival in vitro, in part through increased expression of midkine. A lineage-specific approach might be beneficial for vascular tissue engineering and therapeutic revascularization.


Asunto(s)
Linaje de la Célula/fisiología , Células Madre Embrionarias/citología , Células Endoteliales de la Vena Umbilical Humana/fisiología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/fisiología , Neovascularización Fisiológica/fisiología , Diferenciación Celular , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiología
14.
Breast Care (Basel) ; 8(3): 187-91, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24415968

RESUMEN

BACKGROUND: Omission of axillary lymph node dissection (ALND) is increasingly becoming the new standard of care for patients with sentinel lymph node micrometastases (SNMMs). However, a formidable proportion of patients is afflicted with non-sentinel node (NSN) macrometastatic tumor burden. METHODS: Over 1 decade 5,000 patients underwent sentinel node biopsies (SNB) at 2 certified breast cancer centers in Austria. All available cases of SNMM during this time period were reviewed. Clinical, tumor and lymph node parameters were analyzed using univariate and multivariate analysis to retrieve predictors for further NSN involvement. RESULTS: We identified 216 patients with SNMMs, of whom 181 subsequently underwent ALND. Of the latter patients, 16% (n = 29/181) presented with NSN axillary metastases. ALND revealed NSN macrometastases in 10.5% (n = 19/181) of all the investigated patients, and 66% of the NSN-positive patients (n = 19/29). In 28% (n = 8/29) of the NSN-positive patients, more than 1 macrometastasis was detected. The number of removed sentinel nodes was found to be a significant predictor (p = 0.007) for NSN involvement. CONCLUSION: In this retrospective investigation of breast cancer patients with SNMMs, a substantial proportion exhibited involvement of NSNs. Macrometastases accounted for the largest fraction of NSN tumor burden. Refraining from ALND in the face of SNMMs may entail substantial micro- and macrometastatic tumor burden in the remaining axillary lymph node basin.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA