Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Eur J Immunol ; : e2451207, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38980268

RESUMEN

Tertiary lymphoid structures (TLS) resemble follicles of secondary lymphoid organs and develop in nonlymphoid tissues during inflammation and cancer. Which cell types and signals drive the development of TLS is largely unknown. To investigate early events of TLS development in the lungs, we repeatedly instilled p(I:C) plus ovalbumin (Ova) intranasally. This induced TLS ranging from lymphocytic aggregates to organized and functional structures containing germinal centers. We found that TLS development is independent of FAP+ fibroblasts, alveolar macrophages, or CCL19 but crucially depends on type I interferon (IFN-I). Mechanistically, IFN-I initiates two synergistic pathways that culminate in the development of TLS. On the one hand, IFN-I induces lymphotoxin (LT)α in lymphoid cells, which stimulate stromal cells to produce the B-cell-attracting chemokine CXCL13 through LTßR-signaling. On the other hand, IFN-I is sensed by stromal cells that produce the T-cell-attracting chemokines CXCL9, CXCL10 as well as CCL19 and CCL21 independently of LTßR. Consequently, B-cell aggregates develop within a week, whereas follicular dendritic cells and germinal centers appear after 3 weeks. Thus, sustained production of IFN-I together with an antigen is essential for the induction of functional TLS in the lungs.

2.
Commun Med (Lond) ; 4(1): 5, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38182879

RESUMEN

BACKGROUND: Tertiary lymphoid structures (TLSs) are dense accumulations of lymphocytes in inflamed peripheral tissues, including cancer, and are associated with improved survival and response to immunotherapy in various solid tumors. Histological TLS quantification has been proposed as a novel predictive and prognostic biomarker, but lack of standardized methods of TLS characterization hampers assessment of TLS densities across different patients, diseases, and clinical centers. METHODS: We introduce an approach based on HookNet-TLS, a multi-resolution deep learning model, for automated and unbiased TLS quantification and identification of germinal centers in routine hematoxylin and eosin stained digital pathology slides. We developed HookNet-TLS using n = 1019 manually annotated TCGA slides from clear cell renal cell carcinoma, muscle-invasive bladder cancer, and lung squamous cell carcinoma. RESULTS: Here we show that HookNet-TLS automates TLS quantification across multiple cancer types achieving human-level performance and demonstrates prognostic associations similar to visual assessment. CONCLUSIONS: HookNet-TLS has the potential to be used as a tool for objective quantification of TLS in routine H&E digital pathology slides. We make HookNet-TLS publicly available to promote its use in research.


Tertiary lymphoid structures (TLS) are dense accumulations of immune cells within a cancer. They have been associated with patient survival and treatment effectiveness. Quantification of TLS in cancer microscopy images may therefore aid clinical decision-making. However, no consensus for defining TLS in such images exists leading to inconsistent and variable findings across different labs and studies. We developed a computational tool for automated and objective TLS quantification in cancer images. The tool, called HookNet-TLS, integrates information from multiple image resolutions, which resembles the process of how a pathologist would identify these structures using a microscope. HookNet-TLS detected TLS similarly to trained researchers in three different tumor types. We provided access to HookNet-TLS to facilitate its development and use for TLS assessment in clinical decision-making and research into the role of TLS in cancer.

3.
Cancer Cell ; 41(5): 824-825, 2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-37160101

RESUMEN

Cytotoxic T cells are indispensable for the body's fight against most cancers. In the current issue of Cancer Cell, Gaglia et al. reveal how changes in the tumor tissue architecture creating niches of T cell-B cell interactions may support anti-tumor T cell responses.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Humanos , Microambiente Tumoral , Linfocitos T Citotóxicos , Anticuerpos , Linfocitos B , Neoplasias/terapia
4.
Nat Rev Nephrol ; 19(8): 525-537, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37046081

RESUMEN

Tertiary lymphoid structures (TLSs) are ectopic lymphoid tissues that drive antigen-specific immune responses at sites of chronic inflammation. Unlike secondary lymphoid organs such as lymph nodes, TLSs lack capsules and have their own unique characteristics and functions. The presumed influence of TLSs on the disease course has led to widespread interest in obtaining a better understanding of their biology and function. Studies using single-cell analyses have suggested heterogeneity in TLS composition and phenotype, and consequently, functional correlates with disease progression are sometimes conflicting. The presence of TLSs correlates with a favourable disease course in cancer and infection. Conversely, in autoimmune diseases and chronic age-related inflammatory diseases including chronic kidney disease, the presence of TLSs is associated with a more severe disease course. However, the detailed mechanisms that underlie these clinical associations are not fully understood. To what extent the mechanisms of TLS development and maturation are shared across organs and diseases is also still obscure. Improved understanding of TLS development and function at the cellular and molecular levels may enable the exploitation of these structures to improve therapies for chronic diseases, including chronic kidney disease.


Asunto(s)
Neoplasias , Insuficiencia Renal Crónica , Estructuras Linfoides Terciarias , Humanos , Estructuras Linfoides Terciarias/genética , Estructuras Linfoides Terciarias/patología , Progresión de la Enfermedad , Enfermedad Crónica
5.
Curr Opin Urol ; 31(3): 228-235, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33742986

RESUMEN

PURPOSE OF REVIEW: The aim of this review is to outline characteristics of the renal cell carcinoma (RCC) tumor immune microenvironment (TIME), the potential impact of tumor intrinsic alterations on the TIME and the value of metastatic tissue assessment in this context. RECENT FINDINGS: According to the latest European Association of Urology, European Society for Medical Oncology and National Comprehensive Cancer Network guidelines immune checkpoint inhibition represents a new core treatment strategy in advanced clear cell RCC (ccRCC). Despite its success, the prognosis of many RCC patients remains unsatisfactory most likely because of resistance mechanisms within the TIME. Moreover, most studies assess the primary tumor even though the advanced metastatic disease is targeted. Overall, metastatic RCC has hardly been investigated. First insights into the complexity of the genomic and immune landscape in RCC were recently provided. The functional impact of tumor intrinsic alterations on the TIME has just been described potentially contributing to therapy response in RCC. SUMMARY: The complexity of the RCC TIME and its potential interdependence with tumor intrinsic alterations has only just been recognized. A deeper understanding of the TIME may reveal predictive and prognostic biomarkers long-awaited in RCC, improve RCC patient stratification and could possibly be most instructive if assessed in metastatic tissue.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Biomarcadores de Tumor , Humanos , Neoplasias Renales/terapia , Pronóstico , Microambiente Tumoral
6.
Nat Commun ; 12(1): 769, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33536445

RESUMEN

Some breast tumors metastasize aggressively whereas others remain dormant for years. The mechanism governing metastatic dormancy remains largely unknown. Through high-parametric single-cell mapping in mice, we identify a discrete population of CD39+PD-1+CD8+ T cells in primary tumors and in dormant metastasis, which is hardly found in aggressively metastasizing tumors. Using blocking antibodies, we find that dormancy depends on TNFα and IFNγ. Immunotherapy reduces the number of dormant cancer cells in the lungs. Adoptive transfer of purified CD39+PD-1+CD8+ T cells prevents metastatic outgrowth. In human breast cancer, the frequency of CD39+PD-1+CD8+ but not total CD8+ T cells correlates with delayed metastatic relapse after resection (disease-free survival), thus underlining the biological relevance of CD39+PD-1+CD8+ T cells for controlling experimental and human breast cancer. Thus, we suggest that a primary breast tumor could prime a systemic, CD39+PD-1+CD8+ T cell response that favors metastatic dormancy in the lungs.


Asunto(s)
Antígenos CD/inmunología , Apirasa/inmunología , Neoplasias de la Mama/inmunología , Linfocitos T CD8-positivos/inmunología , Neoplasias Mamarias Experimentales/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Animales , Antígenos CD/metabolismo , Apirasa/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Humanos , Inmunoterapia , Pulmón/inmunología , Pulmón/patología , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/terapia , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones Noqueados , Metástasis de la Neoplasia , Receptor de Muerte Celular Programada 1/metabolismo
7.
Med Image Anal ; 68: 101890, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33260110

RESUMEN

We propose HookNet, a semantic segmentation model for histopathology whole-slide images, which combines context and details via multiple branches of encoder-decoder convolutional neural networks. Concentric patches at multiple resolutions with different fields of view, feed different branches of HookNet, and intermediate representations are combined via a hooking mechanism. We describe a framework to design and train HookNet for achieving high-resolution semantic segmentation and introduce constraints to guarantee pixel-wise alignment in feature maps during hooking. We show the advantages of using HookNet in two histopathology image segmentation tasks where tissue type prediction accuracy strongly depends on contextual information, namely (1) multi-class tissue segmentation in breast cancer and, (2) segmentation of tertiary lymphoid structures and germinal centers in lung cancer. We show the superiority of HookNet when compared with single-resolution U-Net models working at different resolutions as well as with a recently published multi-resolution model for histopathology image segmentation. We have made HookNet publicly available by releasing the source code1 as well as in the form of web-based applications2,3 based on the grand-challenge.org platform.


Asunto(s)
Procesamiento de Imagen Asistido por Computador , Semántica , Mama , Humanos , Redes Neurales de la Computación , Programas Informáticos
8.
Front Immunol ; 12: 793964, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34987518

RESUMEN

Candidate immune biomarkers have been proposed for predicting response to immunotherapy in urothelial cancer (UC). Yet, these biomarkers are imperfect and lack predictive power. A comprehensive overview of the tumor immune contexture, including Tertiary Lymphoid structures (TLS), is needed to better understand the immunotherapy response in UC. We analyzed tumor sections by quantitative multiplex immunofluorescence to characterize immune cell subsets in various tumor compartments in tumors without pretreatment and tumors exposed to preoperative anti-PD1/CTLA-4 checkpoint inhibitors (NABUCCO trial). Pronounced immune cell presence was found in UC invasive margins compared to tumor and stroma regions. CD8+PD1+ T-cells were present in UC, particularly following immunotherapy. The cellular composition of TLS was assessed by multiplex immunofluorescence (CD3, CD8, FoxP3, CD68, CD20, PanCK, DAPI) to explore specific TLS clusters based on varying immune subset densities. Using a k-means clustering algorithm, we found five distinct cellular composition clusters. Tumors unresponsive to anti-PD-1/CTLA-4 immunotherapy showed enrichment of a FoxP3+ T-cell-low TLS cluster after treatment. Additionally, cluster 5 (macrophage low) TLS were significantly higher after pre-operative immunotherapy, compared to untreated tumors. We also compared the immune cell composition and maturation stages between superficial (submucosal) and deeper TLS, revealing that superficial TLS had more pronounced T-helper cells and enrichment of early TLS than TLS located in deeper tissue. Furthermore, superficial TLS displayed a lower fraction of secondary follicle like TLS than deeper TLS. Taken together, our results provide a detailed quantitative overview of the tumor immune landscape in UC, which can provide a basis for further studies.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Ipilimumab/uso terapéutico , Nivolumab/uso terapéutico , Linfocitos T Colaboradores-Inductores/inmunología , Urotelio/metabolismo , Anciano , Antígeno CTLA-4/antagonistas & inhibidores , Diferenciación Celular , Células Cultivadas , Femenino , Técnica del Anticuerpo Fluorescente , Factores de Transcripción Forkhead/metabolismo , Humanos , Inmunoterapia , Activación de Linfocitos , Masculino , Persona de Mediana Edad , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/metabolismo , Estructuras Linfoides Terciarias , Resultado del Tratamiento , Microambiente Tumoral , Neoplasias Urológicas , Urotelio/patología
9.
Front Immunol ; 12: 793992, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35145509

RESUMEN

Lymphoid neogenesis gives rise to tertiary lymphoid structures (TLS) in the periphery of multiple cancer types including muscle invasive bladder cancer (MIBC) where it has positive prognostic and predictive associations. Here, we explored molecular, clinical, and histological data of The Cancer Genome Atlas, as well as the IMvigor210 dataset to study factors associated with TLS development and function in the tumor microenvironment (TME) of MIBC. We also analyzed tumor immune composition including TLS in an independent, retrospective MIBC cohort. We found that the combination of TLS density and tumor mutational burden provides a novel independent prognostic biomarker in MIBC. Gene expression profiles obtained from intratumoral regions that rarely contain TLS in MIBC showed poor correlation with the prognostic TLS density measured in tumor periphery. Tumors with high TLS density showed increased gene signatures as well as infiltration of activated lymphocytes. Intratumoral B-cell and CD8+ T-cell co-infiltration was frequent in TLS-high samples, and such regions harbored the highest proportion of PD-1+TCF1+ progenitor-like T cells, naïve T cells, and activated B cells when compared to regions predominantly infiltrated by either B cells or CD8+ T cells alone. We found four TLS maturation subtypes; however, differences in TLS composition appeared to be dictated by the TME and not by the TLS maturation status. Finally, we identified one downregulated and three upregulated non-immune cell-related genes in TME with high TLS density, which may represent candidates for tumor-intrinsic regulation of lymphoid neogenesis. Our study provides novel insights into TLS-associated gene expression and immune contexture of MIBC and indicates towards the relevance of B-cell and CD8+ T-cell interactions in anti-tumor immunity within and outside TLS.


Asunto(s)
Vasos Linfáticos/metabolismo , Vasos Linfáticos/patología , Músculos/patología , Neovascularización Patológica/etiología , Neovascularización Patológica/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Biomarcadores , Biomarcadores de Tumor , Diagnóstico por Imagen , Susceptibilidad a Enfermedades , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Linfocitos Infiltrantes de Tumor , Densidad Microvascular , Invasividad Neoplásica , Estadificación de Neoplasias , Neovascularización Patológica/diagnóstico por imagen , Pronóstico , Microambiente Tumoral , Neoplasias de la Vejiga Urinaria/etiología , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/mortalidad
10.
Nat Med ; 26(12): 1839-1844, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33046870

RESUMEN

Preoperative immunotherapy with anti-PD1 plus anti-CTLA4 antibodies has shown remarkable pathological responses in melanoma1 and colorectal cancer2. In NABUCCO (ClinicalTrials.gov: NCT03387761 ), a single-arm feasibility trial, 24 patients with stage III urothelial cancer (UC) received two doses of ipilimumab and two doses of nivolumab, followed by resection. The primary endpoint was feasibility to resect within 12 weeks from treatment start. All patients were evaluable for the study endpoints and underwent resection, 23 (96%) within 12 weeks. Grade 3-4 immune-related adverse events occurred in 55% of patients and in 41% of patients when excluding clinically insignificant laboratory abnormalities. Eleven patients (46%) had a pathological complete response (pCR), meeting the secondary efficacy endpoint. Fourteen patients (58%) had no remaining invasive disease (pCR or pTisN0/pTaN0). In contrast to studies with anti-PD1/PD-L1 monotherapy, complete response to ipilimumab plus nivolumab was independent of baseline CD8+ presence or T-effector signatures. Induction of tertiary lymphoid structures upon treatment was observed in responding patients. Our data indicate that combined CTLA-4 plus PD-1 blockade might provide an effective preoperative treatment strategy in locoregionally advanced UC, irrespective of pre-existing CD8+ T cell activity.


Asunto(s)
Ipilimumab/administración & dosificación , Neoplasias/tratamiento farmacológico , Nivolumab/administración & dosificación , Urotelio/patología , Adulto , Anciano , Anticuerpos Monoclonales , Antineoplásicos Inmunológicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/inmunología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/cirugía , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Urotelio/efectos de los fármacos , Urotelio/inmunología , Urotelio/cirugía
11.
Cell Rep ; 29(5): 1236-1248.e7, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31665636

RESUMEN

Sensing of cytoplasmic DNA by cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS) results in production of the dinucleotide cGAMP and consecutive activation of stimulator of interferon genes (STING) followed by production of type I interferon (IFN). Although cancer cells contain supra-normal concentrations of cytoplasmic DNA, they rarely produce type I IFN spontaneously. This suggests that defects in the DNA-sensing pathway may serve as an immune escape mechanism. We find that cancer cells produce cGAMP that is transferred via gap junctions to tumor-associated dendritic cells (DCs) and macrophages, which respond by producing type I IFN in situ. Cancer-cell-intrinsic expression of cGAS, but not STING, promotes infiltration by effector CD8+ T cells and consequently results in prolonged survival. Furthermore, cGAS-expressing cancers respond better to genotoxic treatments and immunotherapy. Thus, cancer-cell-derived cGAMP is crucial to protective anti-tumor CD8+ T cell immunity. Consequently, cancer-cell-intrinsic expression of cGAS determines tumor immunogenicity and makes tumors hot. These findings are relevant for genotoxic and immune therapies for cancer.


Asunto(s)
Neoplasias/inmunología , Nucleotidiltransferasas/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Daño del ADN , Células Dendríticas/metabolismo , Progresión de la Enfermedad , Humanos , Inmunoterapia , Interferón Tipo I/metabolismo , Proteínas de la Membrana , Ratones Endogámicos C57BL , Repeticiones de Microsatélite/genética , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Nucleótidos Cíclicos/metabolismo
13.
Cell ; 177(5): 1330-1345.e18, 2019 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-30982598

RESUMEN

Breast cancer is a heterogeneous disease. Tumor cells and associated healthy cells form ecosystems that determine disease progression and response to therapy. To characterize features of breast cancer ecosystems and their associations with clinical data, we analyzed 144 human breast tumor and 50 non-tumor tissue samples using mass cytometry. The expression of 73 proteins in 26 million cells was evaluated using tumor and immune cell-centric antibody panels. Tumors displayed individuality in tumor cell composition, including phenotypic abnormalities and phenotype dominance. Relationship analyses between tumor and immune cells revealed characteristics of ecosystems related to immunosuppression and poor prognosis. High frequencies of PD-L1+ tumor-associated macrophages and exhausted T cells were found in high-grade ER+ and ER- tumors. This large-scale, single-cell atlas deepens our understanding of breast tumor ecosystems and suggests that ecosystem-based patient classification will facilitate identification of individuals for precision medicine approaches targeting the tumor and its immunoenvironment.


Asunto(s)
Neoplasias de la Mama , Tolerancia Inmunológica , Linfocitos Infiltrantes de Tumor , Macrófagos , Microambiente Tumoral/inmunología , Antígeno B7-H1/inmunología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia sin Enfermedad , Femenino , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/patología , Macrófagos/inmunología , Macrófagos/patología , Proteínas de Neoplasias/inmunología , Tasa de Supervivencia
14.
Front Immunol ; 9: 2660, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30515157

RESUMEN

An important role for tumor infiltrating B lymphocytes (TIL-B) in the immune response to cancer is emerging; however, very little is known about the antigen specificity of antibodies produced in situ. The presence of IgA antibodies in the tumor microenvironment has been noted although their biological functions and clinical significance are unknown. This study used a 91-antigen microarray to examine the IgG and IgA autoantibody repertoires in breast cancer (BC). Tumor and adjacent breast tissue supernatants and plasma from BC patients together with normal breast tissue supernatants and plasma from healthy controls (patients undergoing mammary reduction and healthy blood donors) were analyzed to investigate relationships between autoantibodies and the clinical, histological and immunological features of tumors. Our data show that >84% of the BC samples tested contain autoantibodies to one or more antigens on the array, with ANKRD30BL, COPS4, and CTAG1B being most frequently reactive. Ex vivo TIL-B responses were uncoupled from systemic humoral responses in the majority of cases. A comparison of autoantibody frequencies in supernatants and plasma from patients and controls identified eight antigens that elicit BC-associated autoantibody responses. The overall prevalence of IgG and IgA autoantibodies was similar and while IgG and IgA responses were not linked they did correlate with distinct clinical, pathological and immunological features. Higher levels of ex vivo IgG responses to BC-associated antigens were associated with shorter recurrence-free survival (RFS), HER2 overexpression and lower tumor-infiltrating CD8+ T cell counts. Higher IgA levels were associated with estrogen and progesterone receptor-negative cancers but were not significantly associated with RFS. Furthermore, ex vivo IgA but not IgG autoantibodies reactive to BC-associated antigens were linked with germinal center and early memory B cell maturation and the presence of tertiary lymphoid structures suggesting that these TIL-B are activated in the tumor microenvironment. Overall, our results extend the current understanding of the antigen specificity, the biological and the clinical significance of IgG and IgA autoantibodies produced by BC TIL-B in situ.


Asunto(s)
Especificidad de Anticuerpos , Antígenos de Neoplasias/inmunología , Autoanticuerpos/inmunología , Linfocitos B/inmunología , Neoplasias de la Mama/inmunología , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Adulto , Anciano , Linfocitos B/patología , Neoplasias de la Mama/patología , Femenino , Humanos , Linfocitos Infiltrantes de Tumor/patología , Masculino , Persona de Mediana Edad
15.
Methods Mol Biol ; 1845: 71-86, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30141008

RESUMEN

Tertiary lymphoid structures (TLS) develop in the human tumor microenvironment and correlate with prolonged survival in most cancer types. We recently demonstrated that TLS development follows sequential maturation stages and culminates in the generation of a germinal center (GC) reaction. This maturation process is crucial for the prognostic relevance of TLS in lung and colorectal cancer patients.The mechanisms underlying TLS development in various inflammatory conditions or their functional relevance in tumor immunity are not fully understood. Investigating which cell types and soluble mediators orchestrate lymphoid neogenesis in human tissues requires a method that allows simultaneous detection of multiple markers.Here, we describe a quantitative pathology approach to identify and quantify different TLS maturation stages in combination with other parameters. This approach consists of seven-color immunofluorescence protocol using tyramide signal amplification combined with multispectral microscopy and quantitative data acquisition from histological images.


Asunto(s)
Neoplasias/patología , Estructuras Linfoides Terciarias/patología , Microambiente Tumoral , Biomarcadores , Técnica del Anticuerpo Fluorescente , Centro Germinal/inmunología , Centro Germinal/metabolismo , Centro Germinal/patología , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica/métodos , Microscopía Fluorescente/métodos , Estadificación de Neoplasias , Neoplasias/inmunología , Neoplasias/metabolismo , Programas Informáticos , Estructuras Linfoides Terciarias/inmunología , Estructuras Linfoides Terciarias/metabolismo , Microambiente Tumoral/inmunología
16.
Oncoimmunology ; 7(2): e1378844, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29416939

RESUMEN

Tertiary lymphoid structures (TLS) are associated with favorable outcome in non-metastatic colorectal carcinoma (nmCRC), but the dynamics of TLS maturation and its association with effective anti-tumor immune surveillance in nmCRC are unclear. Here, we hypothesized that not only the number of TLS but also their composition harbors information on recurrence risk in nmCRC. In a comprehensive molecular, tissue, laboratory, and clinical analysis of 109 patients with stage II/III nmCRC, we assessed TLS numbers and degree of maturation in surgical specimens by multi-parameter immunofluorescence of follicular dendritic cell (FDC) and germinal center (GC) markers. TLS formed in most tumors and were significantly more prevalent in highly-microsatellite-instable (MSI-H) and/or BRAF-mutant nmCRC. We could distinguish three sequential TLS maturation stages which were characterized by increasing prevalence of FDCs and mature B-cells: [1] Early TLS, composed of dense lymphocytic aggregates without FDCs, [2] Primary follicle-like TLS, having FDCs but no GC reaction, and [3] Secondary follicle-like TLS, having an active GC reaction. A simple integrated TLS immunoscore reflecting these parameters identified a large subgroup of nmCRC patients with a very low risk of recurrence independently of clinical co-variables such as ECOG performance status, age, stage, and adjuvant chemotherapy. We conclude that (1) mismatch repair and BRAF mutation status are associated with the formation of TLS in nmCRC, (2) TLS formation in nmCRC follows sequential maturation steps, culminating in germinal center formation, and (3) this maturation process harbors important prognostic information on the risk of disease recurrence.

17.
Cancer Res ; 78(5): 1308-1320, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29279354

RESUMEN

In solid tumors, the presence of lymph node-like structures called tertiary lymphoid structures (TLS) is associated with improved patient survival. However, little is known about how TLS develop in cancer, how their function affects survival, and whether they are affected by cancer therapy. In this study, we used multispectral microscopy, quantitative pathology, and gene expression profiling to analyze TLS formation in human lung squamous cell carcinoma (LSCC) and in an experimental model of lung TLS induction. We identified a niche of CXCL13+ perivascular and CXCL12+LTB+ and PD-L1+ epithelial cells supporting TLS formation. We also characterized sequential stages of TLS maturation in LSCC culminating in the formation of germinal centers (GC). In untreated patients, TLS density was the strongest independent prognostic marker. Furthermore, TLS density correlated with GC formation and expression of adaptive immune response-related genes. In patients treated with neoadjuvant chemotherapy, TLS density was similar, but GC formation was impaired and the prognostic value of TLS density was lost. Corticosteroids are coadministered with chemotherapy to manage side effects in LSCC patients, so we evaluated whether they impaired TLS development independently of chemotherapy. TLS density and GC formation were each reduced in chemotherapy-naïve LSCC patients treated with corticosteroids before surgery, compared with untreated patients, a finding that we confirmed in the experimental model of lung TLS induction. Overall, our results highlight the importance of GC formation in TLS during tumor development and treatment.Significance: Corticosteroid treatment during chemotherapy negatively affects the development of tertiary lymphoid structures and abrogates their prognostic value in patients with lung cancer. Cancer Res; 78(5); 1308-20. ©2018 AACR.


Asunto(s)
Corticoesteroides/efectos adversos , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Escamosas/patología , Centro Germinal/patología , Neoplasias Pulmonares/patología , Linfocitos Infiltrantes de Tumor/inmunología , Estructuras Linfoides Terciarias/patología , Anciano , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/inmunología , Proliferación Celular , Femenino , Estudios de Seguimiento , Perfilación de la Expresión Génica , Centro Germinal/efectos de los fármacos , Centro Germinal/inmunología , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Pronóstico , Tasa de Supervivencia , Estructuras Linfoides Terciarias/inducido químicamente , Estructuras Linfoides Terciarias/inmunología , Células Tumorales Cultivadas , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Cancer Epidemiol Biomarkers Prev ; 26(10): 1564-1574, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28768706

RESUMEN

Background: Serum autoantibodies against tumor-associated antigens (TAAs) are detectable in early-stage gastric cancer patients; however, the time point during cancerogenesis when they appear in circulation is still obscure.Methods: In this study, we developed a recombinant antigen microarray and analyzed the prevalence of autoantibodies against 102 TAAs in 829 gastric cancer patients and 929 healthy controls from Caucasian and Asian populations, as well as 100 patients with chronic atrophic gastritis and 775 individuals staged according to different grades of intestinal metaplasia.Results: Six antigens, including CTAG1B/CTAG2, DDX53, IGF2BP2, TP53, and MAGEA3, were predominantly reacting with sera from gastric cancer patients when compared with healthy controls, and the seroreactivity was associated with intestinal-type gastric cancer, but not with patients' Helicobacter pylori status, grade, age, gender, or stage of gastric cancer. We detected gastric cancer-associated seroreactivity in 13% of patients with advanced/severe intestinal metaplasia, which was increased in comparison with mild/moderate intestinal metaplasia (5.3%) and was comparable with that seen in early-stage gastric cancer patients (12%). Moreover, by testing serum samples taken 1 to 9 years before the clinical diagnosis of 18 incident gastric cancer cases, we detected autoantibody responses against several TAAs-SOX2, MYC, BIRC5, IGF2BP1, and MUC1.Conclusions: Our results suggest that humoral immune response against TAAs is generated already during premalignant stages.Impact: Based on the obtained results, cancer-associated autoantibodies might make a valuable contribution to the stratification of high-risk patients with premalignant lesions in the stomach through enhancing the positive predictive power of existing risk models. Cancer Epidemiol Biomarkers Prev; 26(10); 1564-74. ©2017 AACR.


Asunto(s)
Autoanticuerpos/genética , Lesiones Precancerosas/genética , Neoplasias Gástricas/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Prevalencia
19.
Cell ; 169(4): 736-749.e18, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28475899

RESUMEN

Immune cells in the tumor microenvironment modulate cancer progression and are attractive therapeutic targets. Macrophages and T cells are key components of the microenvironment, yet their phenotypes and relationships in this ecosystem and to clinical outcomes are ill defined. We used mass cytometry with extensive antibody panels to perform in-depth immune profiling of samples from 73 clear cell renal cell carcinoma (ccRCC) patients and five healthy controls. In 3.5 million measured cells, we identified 17 tumor-associated macrophage phenotypes, 22 T cell phenotypes, and a distinct immune composition correlated with progression-free survival, thereby presenting an in-depth human atlas of the immune tumor microenvironment in this disease. This study revealed potential biomarkers and targets for immunotherapy development and validated tools that can be used for immune profiling of other tumor types.


Asunto(s)
Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/patología , Neoplasias Renales/inmunología , Neoplasias Renales/patología , Microambiente Tumoral , Humanos , Citometría de Imagen , Tolerancia Inmunológica , Riñón/citología , Macrófagos/inmunología , Macrófagos/patología , Análisis de la Célula Individual , Linfocitos T/inmunología , Linfocitos T/patología
20.
Cancer Immunol Res ; 3(11): 1279-88, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26141620

RESUMEN

In the context of cancer, naïve T cells are insufficiently primed and become progressively dysfunctional. Boosting antitumor responses by blocking PD-1 or CTLA-4 results in durable clinical responses only in a limited proportion of cancer patients, suggesting that other pathways must be targeted to improve clinical efficacy. Our preclinical study in TRAMP mice comparing 14 different immune interventions identified anti-CD40 + IL2/anti-IL2 complexes + IL12Fc as a uniquely efficacious treatment that prevents tolerance induction, promotes priming of sustained, protective tumor-specific CD8(+) T cells, and cures late-stage cancer when given together with adoptively transferred tumor-specific T cells. We propose that improving signals 2 (costimulation) and 3 (cytokines) together with fresh tumor-specific, rather than boosting of dysfunctional preexisting memory, T cells represents a potent therapy for advanced cancer.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Inmunoterapia/métodos , Neoplasias de la Próstata/terapia , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/trasplante , Tolerancia Inmunológica/inmunología , Estimación de Kaplan-Meier , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias de la Próstata/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...