RESUMEN
Food components suppressing small intestinal tumorigenesis are not well-defined partly because of the rarity of this tumor type compared to colorectal tumors. Using Apcmin/+ mice, a mouse model for intestinal tumorigenesis, and antigen-free diet, we report here that food antigens serve this function in the small intestine. By depleting Peyer's patches (PPs), immune inductive sites in the small intestine, we found that PPs have a role in the suppression of small intestinal tumors and are important for the induction of small intestinal T cells by food antigens. On the follicle-associated epithelium (FAE) of PPs, microfold (M) cells pass food antigens from lumen to the dendritic cells to induce T cells. Single-cell RNA-seq (scRNA-seq) analysis of immune cells in PPs revealed a significant impact of food antigens on the induction of the PP T cells and the antigen presentation capacity of dendritic cells. These data demonstrate the role of food antigens in the suppression of small intestinal tumorigenesis by PP-mediated immune cell induction.
Asunto(s)
Células Dendríticas , Neoplasias Intestinales , Intestino Delgado , Ganglios Linfáticos Agregados , Animales , Ratones , Intestino Delgado/inmunología , Intestino Delgado/patología , Neoplasias Intestinales/inmunología , Neoplasias Intestinales/patología , Neoplasias Intestinales/genética , Ganglios Linfáticos Agregados/inmunología , Células Dendríticas/inmunología , Carcinogénesis/inmunología , Antígenos/inmunología , Ratones Endogámicos C57BL , Linfocitos T/inmunología , Presentación de Antígeno/inmunología , Modelos Animales de Enfermedad , AlimentosRESUMEN
Monomorphic epitheliotropic intestinal T-cell lymphoma (MEITL), also known as type II enteropathy-associated T-cell lymphoma, is a rare malignant lymphoma of the extranodal lymphoid tissue derived from interepithelial T lymphocytes. MEITL is a primary intestinal T-cell lymphoma with a challenging diagnosis and aggressive progression, and it can invade other extraintestinal sites. In this study, we report four patients diagnosed with MEITL. All patients presented with abdominal pain, and one patient was admitted because of acute intestinal perforation. Two patients presented with unformed defecation and diarrhea. All patients carried the immunophenotypes CD3, CD7, CD8, CD20, and CD56, and the Ki-67 index ranged 60% to 90%. Three cases were analyzed using next-generation sequencing. One case displayed possibly relevant alterations of CREBBP, NOTCH2, SETD2, and STAT5B, and another case exhibited definite alteration of NOTCH1, possibly relevant alterations of CCND1 and DNMT3A, and potentially relevant alterations of HISTH3B, IGLL5, KMT2C, and KRAS. Different chemotherapy regimens were used, but the prognosis was poor. Hence, we illustrated that because of its low incidence, challenging diagnosis, and difficult treatment, further therapeutic improvements are urgently warranted.
Asunto(s)
Linfoma de Células T Asociado a Enteropatía , Humanos , Linfoma de Células T Asociado a Enteropatía/diagnóstico , Linfoma de Células T Asociado a Enteropatía/tratamiento farmacológico , Linfoma de Células T Asociado a Enteropatía/genética , Linfoma de Células T Asociado a Enteropatía/inmunología , Inmunofenotipificación , Neoplasias Intestinales/diagnóstico , Neoplasias Intestinales/tratamiento farmacológico , Neoplasias Intestinales/genética , Neoplasias Intestinales/inmunología , PronósticoRESUMEN
Approximately 25% of cancers are preceded by chronic inflammation that occurs at the site of tumor development. However, whether this multifactorial oncogenic process, which commonly occurs in the intestines, can be initiated by a specific immune cell population is unclear. Here, we show that an intestinal T cell subset, derived from interleukin-17 (IL-17)-producing helper T (TH17) cells, induces the spontaneous transformation of the intestinal epithelium. This subset produces inflammatory cytokines, and its tumorigenic potential is not dependent on IL-17 production but on the transcription factors KLF6 and T-BET and interferon-γ. The development of this cell type is inhibited by transforming growth factor-ß1 (TGFß1) produced by intestinal epithelial cells. TGFß signaling acts on the pretumorigenic TH17 cell subset, preventing its progression to the tumorigenic stage by inhibiting KLF6-dependent T-BET expression. This study therefore identifies an intestinal T cell subset initiating cancer.
Asunto(s)
Mucosa Intestinal , Factor 6 Similar a Kruppel , Proteínas de Dominio T Box , Células Th17 , Animales , Células Th17/inmunología , Ratones , Proteínas de Dominio T Box/metabolismo , Proteínas de Dominio T Box/genética , Factor 6 Similar a Kruppel/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Transducción de Señal/inmunología , Ratones Endogámicos C57BL , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Ratones Noqueados , Interferón gamma/metabolismo , Interferón gamma/inmunología , Interleucina-17/metabolismo , Interleucina-17/inmunología , Ratones Transgénicos , Proteínas Proto-Oncogénicas/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Neoplasias Intestinales/inmunología , Neoplasias Intestinales/patología , Neoplasias Intestinales/metabolismo , HumanosRESUMEN
Small bowel cancer (SBC) is a rare and aggressive disease with a poor prognosis, necessitating the exploration of novel treatment approaches. This narrative review examines the current evidence on targeted therapy and immunotherapy for SBC, focusing on the two most common subtypes: adenocarcinoma and neuroendocrine tumor. A comprehensive search of PubMed, Scopus, and Google Scholar databases was conducted to identify relevant clinical trials and case reports published in English up to September 2023. The review includes 17 clinical trials and 10 case reports, indicating that targeted therapy and immunotherapy can have the potential to improve survival rates in patients with SBC. Notably, promising targeted medicines include bevacizumab, cetuximab, and trastuzumab, while pembrolizumab and nivolumab show potential as immunotherapies. However, it should be noted that the magnitude of the increase in survival rates with these interventions was small. Further research is needed to determine the optimal combination of targeted therapy and immunotherapy for individual patients with SBC.
Asunto(s)
Inmunoterapia , Neoplasias Intestinales , Terapia Molecular Dirigida , Humanos , Inmunoterapia/métodos , Neoplasias Intestinales/terapia , Neoplasias Intestinales/inmunología , Neoplasias Intestinales/tratamiento farmacológico , Adenocarcinoma/terapia , Adenocarcinoma/inmunología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Intestino Delgado/inmunología , Intestino Delgado/patología , Tumores Neuroendocrinos/terapia , Tumores Neuroendocrinos/inmunología , Tumores Neuroendocrinos/tratamiento farmacológicoRESUMEN
BACKGROUND: Monomorphic epitheliotropic T-cell lymphoma (MEITL) is an aggressive non-Hodgkin lymphoma with a high fatality rate. This study was aimed to explore the clinicopathological and molecular genetic features of MEITL in the Chinese population. METHODS: A retrospective analysis was performed based on the clinical manifestations and pathological features of 20 Chinese MEITL. 9 cases with paired diseased-normal tissues were also analyzed for molecular information by whole-exome sequencing. RESULTS: There were 14 men and 6 women with a median age of 58.5 (28-81) years. 17(17/20) lesions were located in the jejunum or ileum; 13(13/20) cases had ulcers or perforations. Microscopically, except for 1(1/20) case of pleomorphic cells, the monomorphic, middle-sized tumor cells infiltrating into the intestinal epithelial and peripheral intestinal mucosa recess could be seen in the other 19 cases. Immunohistochemistry showed that most of the tumor cells in MEITL were positive for CD3(20/20), CD8(17/20), CD43(19/20), and CD56(15/20), but negative for CD5(20/20). The most frequently mutated genes of these Chinese cases were STAT5B (4/9) and TP53 (4/9), not SETD2(2/9). JAK3 mutations (3/9) were also detected with a high mutated frequency. We demonstrated that mutations of JAK-STAT pathway-related genes and the amplification of Chromosome 9q appeared at the same time in most cases(5/9). CONCLUSIONS: The clinicopathological features were consistent with that in previous western studies, but a special case with pleomorphic cells was found in this study. The co-occurrence of JAK-STAT pathway-related gene mutations and the amplification of Chr9q is a molecular feature of MEITL.
Asunto(s)
Biomarcadores de Tumor/genética , Cromosomas Humanos Par 9 , Amplificación de Genes , Neoplasias Intestinales/diagnóstico , Janus Quinasa 3/genética , Linfoma de Células T/diagnóstico , Mutación , Factor de Transcripción STAT5/genética , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , China , Análisis Mutacional de ADN , Femenino , Predisposición Genética a la Enfermedad , Humanos , Neoplasias Intestinales/genética , Neoplasias Intestinales/inmunología , Neoplasias Intestinales/patología , Linfoma de Células T/genética , Linfoma de Células T/inmunología , Linfoma de Células T/patología , Masculino , Persona de Mediana Edad , Fenotipo , Estudios Retrospectivos , Secuenciación del ExomaRESUMEN
Dendritic cells (DCs), including conventional DCs (cDCs) and plasmacytoid DCs (pDCs), serve as the sentinel cells of the immune system and are responsible for presenting antigen information. Moreover, the role of DCs derived from monocytes (moDCs) in the development of inflammation has been emphasized. Several studies have shown that the function of DCs can be influenced by gut microbes including gut bacteria and viruses. Abnormal changes/reactions in intestinal DCs are potentially associated with diseases such as inflammatory bowel disease (IBD) and intestinal tumors, allowing DCs to be a new target for the treatment of these diseases. In this review, we summarized the physiological functions of DCs in the intestinal micro-environment, their regulatory relationship with intestinal microorganisms and their regulatory mechanism in intestinal diseases.
Asunto(s)
Células Dendríticas/inmunología , Microbioma Gastrointestinal/inmunología , Enfermedades Inflamatorias del Intestino/genética , Neoplasias Intestinales/genética , Intestinos/inmunología , Monocitos/inmunología , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Biomarcadores/metabolismo , Diferenciación Celular , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Citocinas/genética , Citocinas/inmunología , Células Dendríticas/clasificación , Células Dendríticas/citología , Expresión Génica , Humanos , Tolerancia Inmunológica , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/patología , Neoplasias Intestinales/inmunología , Neoplasias Intestinales/microbiología , Neoplasias Intestinales/patología , Intestinos/citología , Intestinos/microbiología , Ratones , Monocitos/citología , Transducción de SeñalRESUMEN
PURPOSE OF REVIEW: Small bowel adenocarcinoma (SBA) is a rare disease, for which few studies have been conducted so far. Therefore, most treatment recommendations have been extrapolated from trials in colorectal cancer. In this review, we revise available data that could improve the management of SBA, with a particular focus on systemic therapy. RECENT FINDINGS: For advanced/irresectable disease, first-line doublet chemotherapy remains standard of care. It is uncertain whether extending treatment to triplet chemotherapy brings added benefit. Pembrolizumab is an accepted treatment modality for mismatch repair-deficient tumors, yet might also be active in microsatellite stable tumors. More trials with immunotherapy are underway. Although there is no place for anti-EGFR monotherapy, the addition of cetuximab to chemotherapy should be investigated further. Two trials suggest an added value of bevacizumab to chemotherapy, yet larger trials are needed to confirm these data. For localized disease, the role of (neo)adjuvant chemotherapy is under investigation. SUMMARY: For decades, patients with SBA have probably been treated suboptimal by basing treatment recommendations on data from colorectal cancer. An effort for SBA-specific trials and/or inclusion of SBA patients in basket trials is of utmost importance in order to improve outcome for these patients.
Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos Inmunológicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Intestinales/tratamiento farmacológico , Intestino Delgado/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Antineoplásicos Inmunológicos/administración & dosificación , Quimioterapia Adyuvante , Ensayos Clínicos Fase II como Asunto , Humanos , Neoplasias Intestinales/inmunología , Neoplasias Intestinales/patología , Intestino Delgado/inmunología , Inhibidores de Proteínas Quinasas/administración & dosificación , Enfermedades Raras/tratamiento farmacológicoAsunto(s)
Neoplasias Intestinales/patología , Intestino Delgado/patología , Linfoma de Células B de la Zona Marginal/patología , Anciano , Femenino , Humanos , Inmunoglobulina A , Cadenas Pesadas de Inmunoglobulina , Cadenas kappa de Inmunoglobulina , Neoplasias Intestinales/inmunología , Intestino Delgado/inmunología , Linfoma de Células B de la Zona Marginal/inmunología , Paraproteinemias/patologíaRESUMEN
Refractory celiac disease type II (RCD II), also referred to as "cryptic" enteropathy-associated T-cell lymphoma (EATL) or "intraepithelial T-cell lymphoma," is a rare clonal lymphoproliferative disorder that arises from innate intraepithelial lymphocytes. RCD II has a poor prognosis and frequently evolves to EATL. The pathogenesis of RCD II is not well understood and data regarding the immunophenotypic spectrum of this disease and underlying genetic alterations are limited. To gain further biological insights, we performed comprehensive immunophenotypic, targeted next-generation sequencing, and chromosome microarray analyses of 11 RCD II cases: CD4-/CD8- (n=6), CD8+ (n=4), and CD4+ (n=1), and 2 of 3 ensuing EATLs. Genetic alterations were identified in 9/11 (82%) of the RCD II cases. All 9 displayed mutations in members of the JAK-STAT signaling pathway, including frequent, recurrent STAT3 (7/9, 78%) and JAK1 (4/9, 44%) mutations, and 9/10 evaluable cases expressed phospho-STAT3. The mutated cases also harbored recurrent alterations in epigenetic regulators (TET2, n=5 and KMT2D, n=5), nuclear factor-κB (TNFAIP3, n=4), DNA damage repair (POT1, n=3), and immune evasion (CD58, n=2) pathway genes. The CD4-/CD8- and other immunophenotypic subtypes of RCD II exhibited similar molecular features. Longitudinal genetic analyses of 4 RCD II cases revealed stable mutation profiles, however, additional mutations were detected in the EATLs, which occurred at extraintestinal sites and were clonally related to antecedent RCD II. Chromosome microarray analysis demonstrated copy number changes in 3/6 RCD II cases, and 1 transformed EATL with sufficient neoplastic burden for informative analysis. Our findings provide novel information about the immunophenotypic and genomic characteristics of RCD II, elucidate early genetic events in EATL pathogenesis, and reveal potential therapeutic targets.
Asunto(s)
Biomarcadores de Tumor/genética , Enfermedad Celíaca/complicaciones , Linfoma de Células T Asociado a Enteropatía/genética , Linfoma de Células T Asociado a Enteropatía/inmunología , Inmunofenotipificación , Neoplasias Intestinales/genética , Neoplasias Intestinales/inmunología , Técnicas de Diagnóstico Molecular , Adulto , Anciano , Preescolar , Análisis Mutacional de ADN , Bases de Datos Factuales , Linfoma de Células T Asociado a Enteropatía/etiología , Femenino , Citometría de Flujo , Reordenamiento Génico , Predisposición Genética a la Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunohistoquímica , Neoplasias Intestinales/etiología , Masculino , Análisis por Micromatrices , Persona de Mediana Edad , Mutación , Ciudad de Nueva York , Fenotipo , Valor Predictivo de las Pruebas , VirginiaRESUMEN
Accumulating evidence has suggested a correlation of tumor infiltrating B cells (TiBcs) and a good prognosis of cancer diseases. In some cases, TiBcs appear to have experienced antigen stimulation since they have undergone class-switching and somatic hypermutation and formed tertiary lymphoid structures around tumors together with T cells. Assuming TiBcs include those that recognize some tumor antigens, we sought to investigate their possible usefulness for cell-mediated immunotherapies. To expand usually small number of TiBcs in vitro, we modified our B cell culture system: we transduced B cells with ERT2-Bach2 so that they grow unlimitedly provided with tamoxifen, IL-21 and our original feeder cells. Such cells differentiate into plasma cells and produce antibodies upon withdrawal of tamoxifen, and further by addition of a Bach2-inhibitor in vitro. As a preliminary experiment, thus expanded splenic B cells expressing a transgenic antigen receptor/antibody against hen egg lysozyme were intravenously injected into mice pre-implanted with B16 melanoma cells expressing membrane-bound HEL in the skin, which resulted in suppression of the growth of B16 tumors and prolonged survival of the recipient mice. To test the usefulness of TiBcs for the immunotherapy, we next used APCmin/+ mice as a model that spontaneously develop intestinal tumors. We cultured TiBcs separated from the tumors of APCmin/+ mice as above and confirmed that the antibodies they produce recognize the APCmin/+ tumor. Repeated injection of such TiBcs into adult APCmin/+ mice resulted in suppression of intestinal tumor growth and elongation of the survival of the recipient mice. Serum antibody from the TiBc-recipient mice selectively bound to an antigen expressed in the tumor of APCmin/+ mice. These data suggest a possibility of the novel individualized cancer immunotherapy, in which TiBcs from surgically excised tumor tissues are expanded and infused into the donor patients.
Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos B/inmunología , Inmunoterapia/métodos , Neoplasias Intestinales/terapia , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Inmunohistoquímica , Interleucinas/farmacología , Neoplasias Intestinales/inmunología , Neoplasias Intestinales/mortalidad , Ratones , Ratones Endogámicos C57BL , Muramidasa/inmunología , Tamoxifeno/farmacologíaRESUMEN
BACKGROUND & AIMS: The reason why small intestinal cancer is rarer than colorectal cancer is not clear. We hypothesized that intraepithelial lymphocytes (IELs), which are enriched in the small intestine, are the closest immune cells to epithelial cells, exclude tumor cells via cell-to-cell contact. METHODS: We developed DPE-green fluorescent protein (DPE-GFP) × adenomatous polyposis coli; multiple intestinal neoplasia (APCmin ) mice, which is a T-cell-reporter mouse with spontaneous intestinal tumors. We visualized the dynamics of IELs in the intestinal tumor microenvironment and the interaction between IELs and epithelial cells, and the roles of cell-to-cell contact in anti-intestinal tumor immunity using a novel in vivo live-imaging system and a novel in vitro co-culture system. RESULTS: In the small intestinal tumor microenvironment, T-cell movement was restricted around blood vessels and the frequency of interaction between IELs and epithelial cells was reduced. Genetic deletion of CD103 decreased the frequency of interaction between IELs and epithelial cells, and increased the number of small intestinal tumors. In the co-culture system, wild-type IELs expanded and infiltrated to intestinal tumor organoids from APCmin mice and reduced the viability of them, which was cell-to-cell contact and CD103 dependent. CONCLUSIONS: The abundance of IELs in the small intestine may contribute to a low number of tumors, although this system may not work in the colon because of the sparseness of IELs. Strategies to increase the number of IELs in the colon or enhance cell-to-cell contact between IELs and epithelial cells may be effective for the prevention of intestinal tumors in patients with a high cancer risk.
Asunto(s)
Antígenos CD/fisiología , Comunicación Celular , Cadenas alfa de Integrinas/fisiología , Mucosa Intestinal/inmunología , Neoplasias Intestinales/prevención & control , Intestino Delgado/inmunología , Linfocitos Intraepiteliales/inmunología , Microambiente Tumoral , Animales , Técnicas de Cocultivo , Femenino , Mucosa Intestinal/citología , Neoplasias Intestinales/inmunología , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Intestino Delgado/patología , Linfocitos Intraepiteliales/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organoides/inmunología , Organoides/patologíaRESUMEN
Gastroenteropancreatic neuroendocrine neoplasms grade 3 (GEP-NENs G3) are rare tumors. These highly aggressive neoplasms are traditionally treated with platinum-based chemotherapy in combination with etoposide. Immune checkpoint proteins such as programmed cell death ligand (PD-L1) may have a role in different cancers allowing them escape the immune system and hence, progress. We aimed to investigate the immunohistochemical expression of PD-L1 in GEP-NEN G3 and evaluate its correlation to clinical parameters. In a cohort of 136 patients, 14 (10%) expressed PD-L1 immunoreactivity; four (3%) patients in the tumor cells and 10 (7%) had immunoreactive immune cells. PD-L1 expression did not correlate to clinical parameters, progression-free survival or overall survival. We conclude that PD-L1 expression is present only in a subset of GEP-NEN G3 patients. Further studies are needed to fully understand the role of PD-L1 in patients with GEP-NEN G3, including the future possibility for treatment with immune checkpoint inhibitors.
Asunto(s)
Antígeno B7-H1/genética , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Intestinales/genética , Tumores Neuroendocrinos/genética , Neoplasias Pancreáticas/genética , Neoplasias Gástricas/genética , Adulto , Anciano , Anciano de 80 o más Años , Antígeno B7-H1/inmunología , Femenino , Humanos , Neoplasias Intestinales/inmunología , Neoplasias Intestinales/patología , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Tumores Neuroendocrinos/inmunología , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Supervivencia sin Progresión , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/patologíaRESUMEN
BACKGROUND & AIMS: Intestinal epithelial cells (IECs) regulate intestinal immune cells, particularly development of T-helper 17 (Th17) cells. Deregulation of this process leads to intestinal inflammation and tumorigenesis, via unknown mechanisms. TANK-binding kinase 1 (TBK1) is expressed by IECs and cells in the innate immune system. We studied the functions of TBK1 in the intestinal immune response and tumorigenesis in mice. METHODS: We performed studies of wild-type mice, mice with conditional disruption of Tbk1 (Tbk1IEC-KO), Tbk1IEC-KO mice crossed with ApcMin/+ mice, and Mt-/- mice crossed with ApcMin/+ mice. Some mice were given intraperitoneal injections of a neutralizing antibody against interleukin 17 (IL17) or IL1ß. Intestine tissues were collected from mice and analyzed by histology, for numbers of adenomas and Th17 cells, and expression of inflammatory cytokines by real-time PCR. IECs were isolated from wild-type and Tbk1IEC-KO mice, stimulated with lipopolysaccharide, co-cultured for with bone marrow-derived macrophages, and analyzed by RNA sequencing and biochemical analyses. RESULTS: Compared to ApcMin/+Tbk1WT mice, ApcMin/+Tbk1IEC-KO mice had significant increases in number and size of intestinal polyps, and significantly more Th17 cells in lamina propria. Administration of an antibody against IL17 reduced the number of intestinal polyps in ApcMin/+Tbk1IEC-KO mice to that observed in ApcMin/+Tbk1WT mice. In culture, TBK1-deficient IECs promoted expression of IL1ß by macrophages, which induced differentiation of naïve CD4+ T cells into Th17 cells. RNA sequencing analysis revealed that the TBK1-deficient IECs had increased expression of metallothionein 1 (MT1), an immune regulator that promotes intestinal inflammation. Intestine tissues from ApcMin/+Mt-/- mice had significant fewer Th17 cells than ApcMin/+Mt+/+ mice, and a significantly lower number of polyps. Analyses of colorectal tumors in the Cancer Genome Atlas found colorectal tumors with high levels of MT1 and IL17 mRNAs to be associated with reduced survival times of patients. CONCLUSIONS: Expression of TBK1 by IECs suppresses expression of MT1 and prevents expression of IL1ß by macrophages and differentiation of Th17 cells, to prevent inflammation and tumorigenesis. Strategies to block this pathway might be developed for colorectal tumorigenesis.
Asunto(s)
Poliposis Adenomatosa del Colon/enzimología , Diferenciación Celular , Transformación Celular Neoplásica/metabolismo , Células Epiteliales/inmunología , Mucosa Intestinal/enzimología , Neoplasias Intestinales/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Células Th17/inmunología , Poliposis Adenomatosa del Colon/inmunología , Poliposis Adenomatosa del Colon/patología , Animales , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/patología , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Células Epiteliales/patología , Genes APC , Humanos , Inmunidad Innata , Inmunidad Mucosa , Interleucina-17/metabolismo , Interleucina-1beta/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Neoplasias Intestinales/inmunología , Neoplasias Intestinales/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Metalotioneína/genética , Metalotioneína/metabolismo , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Células Th17/metabolismoRESUMEN
Cancer stem cells are responsible for tumorigenesis, progression, recurrence and metastasis. Intestinal stem cells (ISCs) are regarded as the origin of intestinal neoplasia. Inflammation also serves an important role in intestinal neoplasia. To explore the molecular mechanisms underlying the inflammationmediated induction of intestinal tumorigenesis, the present study investigated the function of tumor necrosis factor (TNF)α in the malignant transformation of ISCs. NCM460 spheroid (NCM460s) cells with higher expression of stem cell genes, such as Oct4, Nanog, Sox2 and Lgr5, and with a higher ratio of CD133+, were obtained from NCM460 cells in serumfree medium. TNFα accelerated cell proliferation, migration and invasion, induced chemotherapy resistance and the epithelialmesenchymal transition. NFκB and Wnt/ßcatenin pathways were activated in TNFαinduced inflammatory responses, leading to the nuclear translocation of p65 and ßcatenin, as well as promoter activity of NFκB and TCF/LEF transcription factors. It was further demonstrated that TNFαinduced activation of the NFκB and Wnt/ßcatenin signaling pathways, as well as the upregulation of proinflammatory cytokines, were significantly suppressed by p65knockdown. Notably, PDTC, an inhibitor of NFκB signaling, reversed TNFαinduced activation of the NFκB and Wnt/ßcatenin pathways. A similar role was observed for IWP2, an inhibitor of Wnt/ßcatenin signaling. Collectively, these results demonstrated that the NFκB and Wnt/ßcatenin pathways were activated to promote TNFαinduced malignant transformation of ISCs, in which these two pathways crossregulated each other.
Asunto(s)
Células Madre Adultas/patología , Antineoplásicos/farmacología , Transformación Celular Neoplásica/patología , Neoplasias Intestinales/patología , Factor de Necrosis Tumoral alfa/metabolismo , Antineoplásicos/uso terapéutico , Benzotiazoles/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal/inmunología , Técnicas de Silenciamiento del Gen , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/patología , Neoplasias Intestinales/tratamiento farmacológico , Neoplasias Intestinales/inmunología , Prolina/análogos & derivados , Prolina/farmacología , Esferoides Celulares , Tiocarbamatos/farmacología , Factor de Transcripción ReIA/antagonistas & inhibidores , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Regulación hacia Arriba , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/inmunologíaRESUMEN
Tumors have evolved mechanisms to escape anti-tumor immunosurveillance. They limit humoral and cellular immune activities in the stroma and render tumors resistant to immunotherapy. Sensitizing tumor cells to immune attack is an important strategy to revert immunosuppression. However, the underlying mechanisms of immune escape are still poorly understood. Here we discover Indoleamine-2,3-dioxygenase-1 (IDO1)+ Paneth cells in the stem cell niche of intestinal crypts and tumors, which promoted immune escape of colorectal cancer (CRC). Ido1 expression in Paneth cells was strictly Stat1 dependent. Loss of IDO1+ Paneth cells in murine intestinal adenomas with tumor cell-specific Stat1 deletion had profound effects on the intratumoral immune cell composition. Patient samples and TCGA expression data suggested corresponding cells in human colorectal tumors. Thus, our data uncovered an immune escape mechanism of CRC and identify IDO1+ Paneth cells as a target for immunotherapy.
Asunto(s)
Neoplasias Colorrectales/patología , Tolerancia Inmunológica/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Neoplasias Intestinales/patología , Células de Paneth/inmunología , Factor de Transcripción STAT1/fisiología , Animales , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Neoplasias Intestinales/inmunología , Neoplasias Intestinales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones NoqueadosRESUMEN
MMP12 is mainly secreted by macrophages, is involved in macrophage development, and decomposes the extracellular matrix. Herein, we investigated whether macrophages would change in the intestinal tumor microenvironment after MMP12 knockout. ApcMin/+;MMP12-/-mice were obtained by crossbreeding ApcMin/+ mice with MMP12 knockout mice (MMP12-/- mice). The data showed that the number and volume of intestinal tumors were significantly increased in ApcMin/+;MMP12-/- mice compared with ApcMin/+ mice. Additionally, the tumor biomarkers CA19-9, CEA, and ß-catenin appeared relatively early in intestinal tumors in ApcMin/+;MMP12-/- mice. The results demonstrated that knocking out MMP12 accelerated the tumor growth and pathological process. On further investigation of its mechanism, the proportions of M2 macrophages in the spleen and among peritoneal macrophages were significantly up-regulated in ApcMin/+;MMP12-/- mice. Expression of M2 macrophage-related genes was up-regulated in tumor and peritoneal macrophages. The M2-related cytokine levels of IL-4 and IL-13 were increased in the serum of ApcMin/+;MMP12-/-mice. In vitro, bone marrow-derived M2 macrophages were obtained by treating bone marrow cells with IL-4 and IL-13, and these M2 macrophages secreted cytokines being changed. This finding reveals the crucial role of MMP12 in macrophage development and provides a new target for the control of macrophage polarization. Knocking out MMP12 causes intestinal M2 macrophage accumulation in tumor microenvironment, promoting the growth of intestinal tumors in ApcMin/+ mice.
Asunto(s)
Neoplasias Intestinales/inmunología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Macrófagos/patología , Metaloproteinasa 12 de la Matriz/fisiología , Microambiente Tumoral/inmunología , Animales , Citocinas/metabolismo , Femenino , Neoplasias Intestinales/enzimología , Neoplasias Intestinales/patología , Macrófagos/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones NoqueadosRESUMEN
The presence of activated T cells in colorectal cancer tissues is a strong predictor of patient survival. Our previous studies have shown that regulatory T cells (Treg) are able to reduce T cell transendothelial migration in vitro and accumulation of effector T cells in intestinal tumors in vivo in the murine APCMin/+ model for microsatellite stable intestinal tumors. In this study, we investigated the effect of Treg depletion on the density and effector functions of different TCRαß+ and TCRγδ+ T cell populations in intestinal tumors. We used the APCMin/+\DEREG mouse model, which harbor a diphtheria toxin receptor under the control of the FOXP3 promoter, to deplete Treg in tumor bearing mice. We found that the density of conventional TCRαß+CD8αß+ T cells was significantly increased in Treg-depleted tumors in comparison with Treg-proficient tumors. Furthermore, TCRαß+CD8αß+ T cells showed increased proliferation and activation as well as increased Granzyme B and IFN-γ production in Treg-depleted tumors. In sharp contrast, the densities and effector functions of TCRαß+CD8αα+ T cells and TCRγδ+ T cells remained unchanged by Treg depletion. We also documented a distinct population of IL-17A+TNF+ TCRγδ+CD8- T cells in tumors, which were not affected by Treg depletion. We conclude that Treg depletion affects only conventional TCRαß+CD8αß+ T cells in intestinal tumors, while unconventional T cells and T cells in unaffected tissue are not altered. Immunotherapies aimed at depleting Treg from tumors may thus be a viable option for reinvigoration of conventional cytotoxic T cells with a Th1 cytokine profile.
Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Antígenos CD8/inmunología , Linfocitos T CD8-positivos/inmunología , Neoplasias Intestinales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/metabolismo , Femenino , Humanos , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Activación de Linfocitos , Depleción Linfocítica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismoRESUMEN
Small bowel adenocarcinomas (SBAs) are often associated with poor prognosis and have limited therapeutic options. Programmed cell death protein-1 (PD-1)/programmed cell death ligand 1 (PD-L1) pathway blockade is an effective treatment in many microsatellite instability-high (MSI-H) solid tumors. We aimed at investigating PD-L1 and PD-1 expression in non-hereditary, non-ampullary SBAs, associated with celiac disease (CeD), Crohn's disease (CrD), or sporadic, recruited through the Small Bowel Cancer Italian Consortium. We assessed PD-L1 and PD-1 by immunohistochemistry in a series of 121 surgically resected SBAs, including 34 CeD-SBAs, 49 CrD-SBAs, and 38 sporadic SBAs. PD-L1 and PD-1 expression was correlated with several clinico-pathological features, such as the etiology, microsatellite instability status, and tumor-infiltrating lymphocyte (TIL) density. The prevalence of PD-L1 positivity according to combined positive score (CPS) was 26% in the whole cohort of SBAs, with significantly (p = 0.001) higher percentage (35%) in both CeD-SBAs and CrD-SBAs in comparison with sporadic SBAs (5%). CPS ≥ 1 SBAs were significantly (p = 0.013) more frequent in MSI-H cases (41%) than in non-MSI-H ones (18%); however, 15 CPS ≥ 1 microsatellite stable SBAs were also identified. CPS ≥ 1 SBAs showed higher TIL and PD-1+ immune cell density, more frequently medullary histotype, as well as a better outcome in comparison with CPS < 1 cases. This study demonstrates an increased proportion of PD-L1+ cases in both CeD-SBAs and CrD-SBAs in comparison with sporadic SBAs. In addition, the identification of a subset of PD-L1+ microsatellite stable SBAs supports the need to ascertain additional biomarkers of response to immune checkpoint inhibitors along with MSI-H.
Asunto(s)
Adenocarcinoma/patología , Antígeno B7-H1/metabolismo , Neoplasias Intestinales/patología , Intestino Delgado/patología , Adenocarcinoma/etiología , Adenocarcinoma/inmunología , Adulto , Anciano , Biomarcadores de Tumor/análisis , Enfermedad Celíaca/complicaciones , Enfermedad de Crohn/complicaciones , Femenino , Humanos , Neoplasias Intestinales/etiología , Neoplasias Intestinales/inmunología , Linfocitos Infiltrantes de Tumor/patología , Masculino , Inestabilidad de Microsatélites , Persona de Mediana Edad , Estudios RetrospectivosRESUMEN
The tumour microenvironment (TME) of intestinal tumours is highly complex and comprises a network of stromal cells, tumour cells, immune cells and fibroblasts, as well as microorganisms. The tumour location, environmental factors and the tumour cells themselves influence the cells within the TME. Immune cells can destroy tumour cells and are associated with better patient prognosis and response to therapy; however, immune cells are highly plastic and easily influenced to instead promote tumour growth. The interaction between local immune cells and the microbiome can lead to progression or regression of intestinal tumours. In this chapter, we will discuss how tumour development and progression can influence, and be influenced by, the microenvironment surrounding it, focusing on immune and fibroblastic cells, and the intestinal microbiota, particularly in the context of colorectal cancer.
Asunto(s)
Neoplasias Intestinales , Microambiente Tumoral , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Humanos , Neoplasias Intestinales/inmunología , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patologíaRESUMEN
Neuroendocrine neoplasms (NENs) are a group of tumors originating from the neuroendocrine system. They mainly occur in the digestive system and the respiratory tract. It is well-know a strict interaction between neuroendocrine system and inflammation, which can play an important role in NEN carcinogenesis. Inflammatory mediators, which are produced by the tumor microenvironment, can favor cancer induction and progression, and can promote immune editing. On the other hand, a balanced immune system represents a relevant step in cancer prevention through the elimination of dysplastic and cancer cells. Therefore, an inflammatory response may be both pro- and anti-tumorigenic. In this review, we provide an overview concerning the complex interplay between inflammation and gastroenteropancreatic NENs, focusing on the tumorigenesis and clinical implications in these tumors.