Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.009
1.
Gut ; 72(2): 325-337, 2023 02.
Article En | MEDLINE | ID: mdl-35705369

OBJECTIVE: Programmed cell death protein 1 (PD-1) checkpoint inhibition and adoptive cellular therapy have had limited success in patients with microsatellite stable colorectal cancer liver metastases (CRLM). We sought to evaluate the effect of interleukin 10 (IL-10) blockade on endogenous T cell and chimeric antigen receptor T (CAR-T) cell antitumour function in CRLM slice cultures. DESIGN: We created organotypic slice cultures from human CRLM (n=38 patients' tumours) and tested the antitumour effects of a neutralising antibody against IL-10 (αIL-10) both alone as treatment and in combination with exogenously administered carcinoembryonic antigen (CEA)-specific CAR-T cells. We evaluated slice cultures with single and multiplex immunohistochemistry, in situ hybridisation, single-cell RNA sequencing, reverse-phase protein arrays and time-lapse fluorescent microscopy. RESULTS: αIL-10 generated a 1.8-fold increase in T cell-mediated carcinoma cell death in human CRLM slice cultures. αIL-10 significantly increased proportions of CD8+ T cells without exhaustion transcription changes, and increased human leukocyte antigen - DR isotype (HLA-DR) expression of macrophages. The antitumour effects of αIL-10 were reversed by major histocompatibility complex class I or II (MHC-I or MHC-II) blockade, confirming the essential role of antigen presenting cells. Interrupting IL-10 signalling also rescued murine CAR-T cell proliferation and cytotoxicity from myeloid cell-mediated immunosuppression. In human CRLM slices, αIL-10 increased CEA-specific CAR-T cell activation and CAR-T cell-mediated cytotoxicity, with nearly 70% carcinoma cell apoptosis across multiple human tumours. Pretreatment with an IL-10 receptor blocking antibody also potentiated CAR-T function. CONCLUSION: Neutralising the effects of IL-10 in human CRLM has therapeutic potential as a stand-alone treatment and to augment the function of adoptively transferred CAR-T cells.


Carcinoma , Colorectal Neoplasms , Interleukin-10 , Liver Neoplasms , Receptors, Chimeric Antigen , Receptors, Interleukin-10 , Animals , Humans , Mice , Carcinoembryonic Antigen/immunology , Carcinoma/immunology , Carcinoma/secondary , CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/pathology , Immunotherapy, Adoptive , Interleukin-10/antagonists & inhibitors , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Lymphocyte Activation , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Receptors, Interleukin-10/antagonists & inhibitors , Antibodies, Blocking/immunology
2.
J Clin Lab Anal ; 36(5): e24417, 2022 May.
Article En | MEDLINE | ID: mdl-35421271

BACKGROUND: Autophagy plays a vital role in the progression of the tumor. We aimed to investigate the expression, prognostic value, and immune infiltration of autophagy-related genes in oral carcinoma via bioinformatics analysis. METHODS: The microarray datasets (GSE146483 and GSE23558) of oral carcinoma were downloaded from Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) between normal and diseased groups were identified by the Limma package. The screened autophagy-related gene was further validated by the human protein atlas (HPA) database, TCGA database, and GSE78060 dataset. RESULTS: A total of 18 upregulated (top 10: EGFR, TNF, FADD, AURKA, E2F1, CHEK1, BRCA1, BIRC5, EIF2AK2, and CSF2) and 31 downregulated (top 10: MAP1LC3A, PARK2, AGT, IGF1, TP53INP1, CXCL12, IKBKB, SESN1, ULK2, and RRAGD) autophagy-related (DEGs) were identified, and FADD was found to be related to the prognosis of oral cancer patients. Gene set enrichment analysis indicated that FADD-associated genes were significantly enriched in immune-related pathways. Moreover, correlation analysis revealed that FADD expression was associated with immune infiltrates. Upregulation of FADD is associated with poor survival and immune infiltrates in oral cancer. CONCLUSION: We speculated that FADD is involved in the immune regulation of oral cancer, as well as autophagy.


Autophagy , Carcinoma , Mouth Neoplasms , Autophagy/genetics , Biomarkers, Tumor/genetics , Carcinoma/genetics , Carcinoma/immunology , Gene Expression Regulation, Neoplastic , Humans , Mouth Neoplasms/genetics , Mouth Neoplasms/immunology , Prognosis
3.
Anticancer Res ; 42(3): 1629-1634, 2022 Mar.
Article En | MEDLINE | ID: mdl-35220261

BACKGROUND/AIM: This study aimed to clarify the impact of proton pump inhibitors (PPIs) on oncological outcomes in patients who received pembrolizumab for advanced urothelial carcinoma (UC). PATIENTS AND METHODS: Forty advanced UC patients treated with pembrolizumab were retrospectively reviewed and divided into two groups (PPI: 15 patients; without PPI: 25 patients). Tumor response and survival were compared between these groups. The factors associated with survival were also investigated. RESULTS: The objective response rate was significantly lower in the group with PPIs compared with the group without PPIs. Both progression-free survival (PFS) and overall survival (OS) were significantly shorter in the group with PPIs than in the group without PPIs. The use of PPIs was a significant predictor of poor PFS and OS in multivariate analysis. CONCLUSION: The use of PPIs was negatively associated with tumor response and survival in patients with advanced UC treated with pembrolizumab.


Antibodies, Monoclonal, Humanized/therapeutic use , Carcinoma/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Proton Pump Inhibitors/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/adverse effects , Carcinoma/immunology , Carcinoma/mortality , Carcinoma/pathology , Female , Humans , Immune Checkpoint Inhibitors/adverse effects , Male , Progression-Free Survival , Proton Pump Inhibitors/adverse effects , Retrospective Studies , Risk Assessment , Risk Factors , Time Factors , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology
4.
Sci Rep ; 12(1): 1222, 2022 01 24.
Article En | MEDLINE | ID: mdl-35075167

The significance of N6-methyladenosine (m6A) RNA modifications in the progression of breast cancer (BC) has been recognised. However, their potential role and mechanism of action in the tumour microenvironment (TME) and immune response has not been demonstrated. Thus, the role of m6A regulators and their downstream target gene components in BC remain to be explored. In this study, we used a series of bioinformatics methods and experiments to conduct exploratory research on the possible role of m6A regulators in BC. First, two regulatory modes of immune activation and inactivation were determined by tumour classification. The TME, immune cell infiltration, and gene set variation analysis results confirmed the reliability of this pattern. The prognostic model of the m6A regulator was established by the least absolute shrinkage and selection operator and univariate and multivariate Cox analyses, with the two regulators most closely related to survival verified by real-time quantitative reverse transcription polymerase chain reaction. Next, the prognostic m6A regulator identified in the model was crossed with the differential copy number of variant genes in invasive BC (IBC), and it was determined that YTHDF1 was a hub regulator. Subsequently, single-cell analysis revealed the expression patterns of m6A regulators in different IBC cell populations and found that YTHDF1 had significantly higher expression in immune-related IBC cells. Therefore, we selected the intersection of the BC differential expression gene set and the differential expression gene set of a cell line with knocked-down YTHDF1 in literature to identify downstream target genes of YTHDF1, in which we found IFI6, EIR, and SPTBN1. A polymerase chain reaction was conducted to verify the results. Finally, we confirmed the role of YTHDF1 as a potential prognostic biomarker through pan-cancer analysis. Furthermore, our findings revealed that YTHDF1 can serve as a new molecular marker for BC immunotherapy.


Adenosine/analogs & derivatives , Breast Neoplasms/metabolism , Carcinoma/metabolism , RNA/metabolism , Tumor Microenvironment , Adenosine/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Carcinoma/drug therapy , Carcinoma/immunology , Carcinoma/mortality , Case-Control Studies , Humans , Lymphocytes, Tumor-Infiltrating , Molecular Targeted Therapy , Prognosis , RNA-Binding Proteins/metabolism
5.
Cells ; 11(1)2022 01 05.
Article En | MEDLINE | ID: mdl-35011737

David Mason started his research career at a time when lymphoma diagnosis was based primarily on cellular morphology, clinical symptoms and special cytochemical stains using formalin fixed tissue sections. There were occasions, however, where the morphology was unhelpful, such as in the case of anaplastic or poorly differentiated tumours, where a distinction between lymphoma and a non-haematopoietic tumour was often problematical. Accurate diagnosis became even more important with the developments in the clinical staging of lymphoma and the availability of more effective treatments. One way forward to improve diagnosis was to use immunohistochemistry to study the antigens expressed by the tumor cells.


Carcinoma/pathology , Cooperative Behavior , Lymphoma/pathology , Carcinoma/diagnosis , Carcinoma/immunology , Humans , Leukemia/diagnosis , Leukemia/immunology , Lymphoma/diagnosis , Lymphoma/immunology
6.
Gastroenterology ; 162(3): 907-919.e10, 2022 03.
Article En | MEDLINE | ID: mdl-34863788

BACKGROUND & AIMS: Owing to the high load of immunogenic frameshift neoantigens, tumors arising in individuals with Lynch syndrome (LS), the most common inherited colorectal cancer (CRC) syndrome, are characterized by a pronounced immune infiltration. However, the immune status of normal colorectal mucosa in LS is not well characterized. We assessed the immune infiltrate in tumor-distant normal colorectal mucosa from LS CRC patients, sporadic microsatellite-unstable (MSI) and microsatellite-stable (MSS) CRC patients, and cancer-free LS carriers. METHODS: CD3-positive, FOXP3-positive, and CD8-positive T cells were quantified in, respectively, 219, 233, and 201 formalin-fixed paraffin-embedded (FFPE) normal colonic mucosa tissue sections from CRC patients and cancer-free LS carriers and 26, 22, and 19 LS CRCs. CD3-positive T cells were also quantified in an independent cohort of 97 FFPE normal rectal mucosa tissue sections from LS carriers enrolled in the CAPP2 clinical trial. The expression of 770 immune-relevant genes was analyzed in a subset of samples with the use of the NanoString nCounter platform. RESULTS: LS normal mucosa specimens showed significantly elevated CD3-, FOXP3-, and CD8-positive T-cell densities compared with non-LS control specimens. Gene expression profiling and cluster analysis revealed distinct immune profiles in LS carrier mucosa with and without cancer manifestation. Long-term follow-up of LS carriers within the CAPP2 trial found a correlation between mucosal T-cell infiltrate and time to subsequent tumor occurrence. CONCLUSIONS: LS carriers show elevated mucosal T-cell infiltration even in the absence of cancer. The normal mucosa immune profile may be a temporary or permanent tumor risk modifier in LS carriers.


Carcinoma/immunology , Colon/immunology , Colorectal Neoplasms, Hereditary Nonpolyposis/immunology , Intestinal Mucosa/immunology , Rectum/immunology , T-Lymphocytes/metabolism , Adult , Aged , Aged, 80 and over , CD3 Complex/metabolism , CD8-Positive T-Lymphocytes/pathology , Carcinoma/genetics , Carcinoma/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , DNA-Binding Proteins/genetics , Female , Forkhead Transcription Factors/metabolism , Heterozygote , Humans , Intestinal Mucosa/pathology , Lymphocyte Count , Male , Microsatellite Instability , Middle Aged , Mismatch Repair Endonuclease PMS2/genetics , MutL Protein Homolog 1/genetics , MutS Homolog 2 Protein/genetics , T-Lymphocytes/pathology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Transcriptome , Young Adult
7.
J Immunol Res ; 2021: 1589864, 2021.
Article En | MEDLINE | ID: mdl-34926701

BACKGROUND: The lncRNA BACE1-AS was identified as a plasma molecular marker in the early diagnosis of Alzheimer's disease, but its role in tumors remains poorly defined. METHODS: The expression patterns, genomic mutation, and prognostic significance of BACE1-AS in pan-cancers were compared by analyzing 32 types of tumors from The Cancer Genome Atlas and cBioPortal databases. The relationships between BACE1-AS expression levels and the degree of immune cell infiltration, immune components, and immune-related genes were explored. The possible molecular mechanisms of BACE1-AS in tumors were explored using gene set enrichment analysis (GSEA). Finally, the role of BACE1-AS in hepatocellular carcinoma was confirmed via quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS: BACE1-AS expression levels were significantly upregulated in LIHC, GBM, KIRC, CHOL, STAD, KICH, COAD, and PRAD. Higher expression levels of BACE1-AS were associated with worse overall survival in patients with HNSC and LIHC, while the opposite was found in PCPG and THCA. The overall mutation rate of BACE1-AS in pan-cancer was only approximately 0.9%, and it occurred mainly in uveal melanoma and uterine carcinoma. Generally, BACE1-AS expression was negatively correlated with the immune microenvironment. BACE1-AS expression was mainly related to naïve B cells, activated memory CD4 T cells, monocytes, M1 macrophages, M2 macrophages, and resting mast cells. The potential mechanisms of BACE1-AS in tumors were mainly via regulating the activities of B cell-mediated immunity, immune response regulating cell surface receptor signaling, RNA binding in posttranscriptional gene silencing, B cell receptor signaling pathways, and immune receptor activity. Finally, the qRT-PCR results confirmed that the expression levels of BACE1-AS in hepatocellular carcinoma cell lines were upregulated. CONCLUSIONS: Overall, our results suggest that BACE1-AS is associated with the expression, prognosis, and rate of immune cell infiltration of most tumors. Thus, BACE1-AS may be a potential target for immunotherapies aimed at improving cancer patient outcomes.


Biomarkers, Tumor/metabolism , Carcinogenesis/genetics , Carcinoma/genetics , RNA, Long Noncoding/metabolism , Adult , Aged , Carcinogenesis/immunology , Carcinoma/immunology , Carcinoma/mortality , Carcinoma/pathology , Datasets as Topic , Female , Gene Expression Regulation, Neoplastic/immunology , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Risk Assessment/methods , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
8.
Front Immunol ; 12: 788431, 2021.
Article En | MEDLINE | ID: mdl-34970268

Background: Uterine corpus endometrial carcinoma (UCEC) is a gynecological malignant tumor with low survival rate and poor prognosis. The traditional clinicopathological staging is insufficient to estimate the prognosis of UCEC. It is necessary to select a more effective prognostic signature of UCEC to predict the prognosis and immunotherapy effect of UCEC. Methods: CIBERSORT and weighted correlation network analysis (WGCNA) algorithms were combined to screen modules related to regulatory T (Treg) cells. Subsequently, univariate, least absolute shrinkage and selection operator (LASSO), and multivariate Cox regression analyses were used to identify the genes in key modules. The difference in overall survival (OS) between high- and low-risk patients was analyzed by Kaplan-Meier analysis. The Tregs-related risk signature (TRRS) was screened by uni- and multivariate Cox analyses. Afterward, we analyzed the expression difference of TRRS and verified its ability to predict the prognosis of UCEC and the effect of immunotherapy. Results: Red module has the highest correlation with Tregs among all clustered modules. Pathways enrichment indicated that the related processes of UCEC were primarily associated to the immune system. Eight genes (ZSWIM1, NPRL3, GOLGA7, ST6GALNAC4, CDC16, ITPK1, PCSK4, and CORO1B) were selected to construct TRRS. We found that this TRRS is a significantly independent prognostic factor of UCEC. Low-risk patients have higher overall survival than high-risk patients. The immune status of different groups was different, and tumor-related pathways were enriched in patients with higher risk score. Low-risk patients are more likely take higher tumor mutation burden (TMB). Meanwhile, they are more sensitive to chemotherapy than patients with high-risk score, which indicated a superior prognosis. Immune checkpoints such as PD-1, CTLA4, PD-L1, and PD-L2 all had a higher expression level in low-risk group. TRRS expression really has a relevance with the sensitivity of UCEC patients to chemotherapeutic drugs. Conclusion: We developed and validated a TRRS to estimate the prognosis and reflect the immune status of UCEC, which could accurately assess the prognosis of patients with UCEC and supply personalized treatments for them.


Biomarkers, Tumor/genetics , Carcinoma/genetics , Endometrial Neoplasms/genetics , Gene Expression Profiling , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes, Regulatory/immunology , Transcriptome , Tumor Microenvironment/immunology , Carcinoma/drug therapy , Carcinoma/immunology , Carcinoma/mortality , Case-Control Studies , Clinical Decision-Making , Databases, Genetic , Decision Support Techniques , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/immunology , Endometrial Neoplasms/mortality , Female , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genome-Wide Association Study , Humans , Immunotherapy , Nomograms , Predictive Value of Tests , Prognosis , Reproducibility of Results , Risk Assessment , Risk Factors
9.
Anticancer Res ; 41(11): 5749-5759, 2021 Nov.
Article En | MEDLINE | ID: mdl-34732448

BACKGROUND/AIM: Systemic immune-inflammation index (SII) predicts survival of patients with various malignancies. This study explored the prognostic value of SII in metastatic urothelial carcinoma (MUC) subjects. PATIENTS AND METHODS: We evaluated 181 consecutive MUC patients treated with first-line platinum-based therapy. Karnofsky performance status <80% and visceral metastasis were present in 18.2% and 46.4% of patients, respectively. SII was based on platelet × neutrophil/lymphocyte counts. Study population was dichotomized by median into high and low SII groups before the initiation of chemotherapy and at week 6. Progression-free survival (PFS) and overall survival (OS) were estimated by the Kaplan-Meier method and compared with the log-rank test. RESULTS: At median follow-up of 9.6 months, 174 patients experienced disease progression and 173 died. Patients with low SII at baseline and at week 6 had significantly better PFS (HR=0.58; p=0.0002 and HR=0.55; p<0.0001) and OS (HR=0.54; p<0.0001 and HR=0.54; p<0.0001) compared to patients with high SII. Independent prognostic value of SII was confirmed in a multivariate analysis. CONCLUSION: High SII before chemotherapy that persists at week 6 negatively affects survival. SII at baseline can be used in the stratification of patients within clinical trials and in clinical practice.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blood Platelets , Carboplatin/therapeutic use , Carcinoma/drug therapy , Cisplatin/therapeutic use , Lymphocytes , Neutrophils , Urinary Bladder Neoplasms/drug therapy , Urothelium/drug effects , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carboplatin/adverse effects , Carcinoma/immunology , Carcinoma/mortality , Carcinoma/secondary , Cisplatin/adverse effects , Female , Humans , Lymphocyte Count , Male , Middle Aged , Platelet Count , Predictive Value of Tests , Progression-Free Survival , Retrospective Studies , Risk Assessment , Risk Factors , Slovakia/epidemiology , Time Factors , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology , Urothelium/immunology , Urothelium/pathology , White People , Young Adult
10.
Sci Rep ; 11(1): 21314, 2021 10 29.
Article En | MEDLINE | ID: mdl-34716378

The release of Hsp70 chaperone from tumor cells is found to trigger the full-scale anti-cancer immune response. Such release and the proper immune reaction can be induced by the delivery of recombinant Hsp70 to a tumor and we sought to explore how the endogenous Hsp70 can be transported to extracellular space leading to the burst of anti-cancer activity. Hsp70 transport mechanisms were studied by analyzing its intracellular tracks with Rab proteins as well as by using specific inhibitors of membrane domains. To study Hsp70 forms released from cells we employed the assay consisting of two affinity chromatography methods. Hsp70 content in culture medium and extracellular vesicles (EVs) was measured with the aid of ELISA. The properties and composition of EVs were assessed using nanoparticle tracking analysis and immunoblotting. The activity of immune cells was studied using an assay of cytotoxic lymphocytes, and for in vivo studies we employed methods of affinity separation of lymphocyte fractions. Analyzing B16 melanoma cells treated with recombinant Hsp70 we found that the chaperone triggered extracellular transport of its endogenous analog in soluble and enclosed in EVs forms; both species efficiently penetrated adjacent cells and this secondary transport was corroborated with the strong increase of Natural Killer (NK) cell toxicity towards melanoma. When B16 and CT-26 colon cancer cells before their injection in animals were treated with Hsp70-enriched EVs, a powerful anti-cancer effect was observed as shown by a two-fold reduction in tumor growth rate and elevation of life span. We found that the immunomodulatory effect was due to the enhancement of the CD8-positive response and anti-tumor cytokine accumulation; supporting this there was no delay in CT-26 tumor growth when Hsp70-enriched EVs were grafted in nude mice. Importantly, pre-treatment of B16 cells with Hsp70-bearing EVs resulted in a decline of arginase-1-positive macrophages, showing no generation of tumor-associated macrophages. In conclusion, Hsp70-containing EVs generated by specifically treated cancer cells give a full-scale and effective pattern of anti-tumor immune responses.


Adaptive Immunity , Extracellular Vesicles , HSP70 Heat-Shock Proteins/pharmacology , Animals , Carcinoma/immunology , Cell Line, Tumor , Colonic Neoplasms/immunology , HEK293 Cells , Humans , Killer Cells, Natural/immunology , Melanoma, Experimental/immunology , Mice
11.
Int J Mol Sci ; 22(17)2021 Sep 02.
Article En | MEDLINE | ID: mdl-34502435

Locally advanced or metastatic urothelial bladder cancer (a/m UBC) is currently treated using platinum-based combination chemotherapy. Immune checkpoint inhibitors (ICIs) are the preferred second-line treatment options for cisplatin-eligible a/m UBC patients and as first-line options in cisplatin-ineligible settings. However, the response rates for ICI monotherapy are modest (~20%), which necessitates the exploration of alternative strategies. Dysregulated activation of fibroblast growth factor receptor (FGFR) signaling enhances tumor proliferation, survival, invasion, angiogenesis, and immune evasion. The recent U.S. Food and Drug Administration approval of erdafitinib and the emergence of other potent and selective FGFR inhibitors (FGFRis) have shifted the treatment paradigm for patients with a/m UBC harboring actionable FGFR2 or FGFR3 genomic alterations, who often have a minimal-to-modest response to ICIs. FGFRi-ICI combinations are therefore worth exploring, and their preliminary response rates and safety profiles are promising. In the present review, we summarize the impact of altered FGFR signaling on a/m UBC tumor evolution, the clinical development of FGFRis, the rationale for FGFRi-ICI combinations, current trials, and prospective research directions.


Antineoplastic Agents/therapeutic use , Carcinoma/drug therapy , Fibroblast Growth Factors/antagonists & inhibitors , Tumor Microenvironment/drug effects , Urinary Bladder Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols , Carcinoma/immunology , Humans , Immune Checkpoint Inhibitors/therapeutic use , Urinary Bladder Neoplasms/immunology
12.
Int J Med Sci ; 18(14): 3249-3260, 2021.
Article En | MEDLINE | ID: mdl-34400894

Dendritic cell (DC)-based immunotherapy has been a promising strategy for colon cancer therapy, but the efficacy of dendritic cell vaccines is in part limited by immunogenicity of loaded antigens. In this study, we aimed to identify a putative tumor antigen that can generate or enhance anti-tumor immune responses against colon cancer. CD44+ colon cancer stem cells (CCSCs) were isolated from mouse colorectal carcinoma CT-26 cell cultures and induced to form defective ribosomal products-containing autophagosome-rich blebs (DRibbles) by treatment with rapamycin, bortezomib, and ammonium chloride. DRibbles were characterized by western blot and transmission electron microscopy. DCs generated from the mice bone marrow monocytes were cocultured with DRibbles, then surface markers of DCs were analyzed by flow cytometry. Meanwhile, the efficacy of DRibble-DCs was examined in vivo. Our results showed that CCSC-derived DRibbles upregulated CD80, CD86, major histocompatibility complex (MHC)-I, and MHC-II on DCs and induced proliferation of mouse splenic lymphocytes and CD8+ T cells. In a model of colorectal carcinoma using BALB/c mice with robust tumor growth and mortality, DC vaccine pulsed with CCSC-derived DRibbles suppressed tumor growth and extended survival. A lactate dehydrogenase test indicated a strong cytolytic activity of cytotoxic T-cells derived from mice vaccinated with CCSC-derived DRibbles against CT-26 cells. Furthermore, flow cytometry analyses showed that the percentages of IFN-γ-producing CD8+ T-cells were increased in SD-DC group compare with the other groups. These findings provide a rationale for novel immunotherapeutic anti-tumor approaches based on DRibbles derived from colon cancer stem cells.


Cancer Vaccines/administration & dosage , Carcinoma/therapy , Colorectal Neoplasms/therapy , Neoplastic Stem Cells/immunology , Ammonium Chloride/pharmacology , Animals , Autophagosomes/drug effects , Autophagosomes/immunology , Bortezomib/pharmacology , Cancer Vaccines/immunology , Carcinoma/immunology , Carcinoma/pathology , Cell Line, Tumor , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Dendritic Cells/immunology , Disease Models, Animal , Female , Humans , Immunogenicity, Vaccine , Mice , Neoplastic Stem Cells/drug effects , Primary Cell Culture , Sirolimus/pharmacology , T-Lymphocytes, Cytotoxic/immunology
13.
Front Immunol ; 12: 692859, 2021.
Article En | MEDLINE | ID: mdl-34276684

Background: Tumor-infiltrating lymphocytes (TILs) are considered a manifestation of the host immune response against cancer and tertiary lymphoid structures (TLS) may contribute to lymphocytes recruitment. Both of them have been reported as potential prognostic parameters in some human malignancies. However, the roles of TILs, TLS, and their correlation in Epstein-Barr Virus-associated gastric carcinoma (EBVaGC) and EBV-negative gastric carcinoma (EBVnGC) are largely unknown. Methods: To observe the correlation among TILs, TLS, and clinicopathological characteristics and their prognostic significance in EBVaGC and EBVnGC, respectively. TILs and TLS were assessed by morphology and/or immunohistochemistry, and accompanied by clinicopathological analysis from 846 gastric cancer patients in multiple institutions. Results: Forty-two (5.0%) cases of EBVaGC and 804 cases of EBVnGC were identified by in situ hybridization, respectively. For EBVnGC, higher TILs grade was correlated with TLS-present. EBVnGC patients with high TILs grade and TLS-present exhibited survival benefits. TILs (P = 0.001) and TLS (P = 0.003), especially TILs & TLS (P < 0.001) were independent prognostic factors in EBVnGC. A nomogram was constructed and validated for predicting the probability of overall survival and performed well with a good calibration. No significant prognostic value was detected in EBVaGC. Conclusion: TILs and TLS, especially TILs & TLS were promising prognostic indicators for overall survival in EBVnGC. TILs and TLS were highly overlapping in their extent and prognostic abilities, and may be considered as a coindicator of prognosis of gastric cancer. The evaluations of TILs and TLS are simple and can be assessed routinely in pathological diagnosis.


Carcinoma/diagnosis , Decision Support Techniques , Herpesvirus 4, Human/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Nomograms , Stomach Neoplasms/diagnosis , Tertiary Lymphoid Structures/immunology , Tumor Microenvironment/immunology , Carcinoma/immunology , Carcinoma/therapy , Carcinoma/virology , China , Female , Herpesvirus 4, Human/pathogenicity , Host-Pathogen Interactions , Humans , Lymphocytes, Tumor-Infiltrating/virology , Male , Middle Aged , Predictive Value of Tests , Prognosis , Reproducibility of Results , Risk Assessment , Risk Factors , Stomach Neoplasms/immunology , Stomach Neoplasms/therapy , Stomach Neoplasms/virology , Tertiary Lymphoid Structures/virology
14.
Front Immunol ; 12: 634741, 2021.
Article En | MEDLINE | ID: mdl-34220801

Immune checkpoint inhibitors (ICIs) have changed the treatment paradigm of metastatic urothelial carcinoma (mUC), a dominant type of bladder cancer (BC). Previous studies have shown an association between gene mutations in the DNA damage response (DDR) pathway and the immunotherapy response in mUC but have neglected the effect of the activation level of the DDR pathway on the ICI response in mUC. A published immunotherapy cohort with genome, transcriptome and survival data for 348 mUC patients was used. An external cohort (The Cancer Genome Atlas Bladder Cancer) and the GSE78220 cohort were used for validation. The activation level of the DDR pathway was quantified using single-sample gene set enrichment analysis (ssGSEA). Further analysis on the genome, immunogenicity, and the immune microenvironment was conducted using the DDR ssGSEA enrichment score-high (DSSH) group and the DDR ssGSEA enrichment score-low (DSSL) group. In the mUC cohorts, the DSSH group was associated with longer overall survival times (P=0.026; Hazard ratio=0.67; 95%CI: 0.46-0.95). The DSSH group was also associated with higher tumor mutation burden, neoantigen load, immune-activated cell patterns, and immune-related gene expression levels. The GSEA results indicated an immune activation state in DSSH group, which correlated with a down-regulation in the transforming growth factor ß receptor signaling pathway. Our study suggests that the activation level of the DDR pathway may be a novel predictive marker for immunotherapy efficacy in patients with mUC.


Carcinoma/drug therapy , DNA Damage , DNA Repair , Immune Checkpoint Inhibitors/therapeutic use , Transforming Growth Factor beta/genetics , Urinary Bladder Neoplasms/drug therapy , Urothelium/drug effects , Carcinoma/genetics , Carcinoma/immunology , Carcinoma/metabolism , Databases, Genetic , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Metastasis , Signal Transduction , Transcriptome , Transforming Growth Factor beta/metabolism , Treatment Outcome , Tumor Microenvironment , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/metabolism , Urothelium/metabolism , Urothelium/pathology
15.
Front Immunol ; 12: 690201, 2021.
Article En | MEDLINE | ID: mdl-34220848

Ovarian carcinomas (OCs) are poorly immunogenic and immune checkpoint inhibitors (ICIs) have offered a modest benefit. In this study, high CD3+ T-cells and CD163+ tumor-associated macrophages (TAMs) densities identify a subgroup of immune infiltrated high-grade serous carcinomas (HGSCs) with better outcomes and superior response to platinum-based therapies. On the contrary, in most clear cell carcinomas (CCCs) showing poor prognosis and refractory to platinum, a high TAM density is associated with low T cell frequency. Immune infiltrated HGSC are characterized by the 30-genes signature (OC-IS30) covering immune activation and IFNγ polarization and predicting good prognosis (n = 312, TCGA). Immune infiltrated HGSC contain CXCL10 producing M1-type TAM (IRF1+pSTAT1Y701+) in close proximity to T-cells. A fraction of these M1-type TAM also co-expresses TREM2. M1-polarized TAM were barely detectable in T-cell poor CCC, but identifiable across various immunogenic human cancers. Single cell RNA sequencing data confirm the existence of a tumor-infiltrating CXCL10+IRF1+STAT1+ M1-type TAM overexpressing antigen processing and presentation gene programs. Overall, this study highlights the clinical relevance of the CXCL10+IRF1+STAT1+ macrophage subset as biomarker for intratumoral T-cell activation and therefore offers a new tool to select patients more likely to respond to T-cell or macrophage-targeted immunotherapies.


Carcinoma/metabolism , Chemokine CXCL10/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms, Cystic, Mucinous, and Serous/metabolism , Ovarian Neoplasms/metabolism , Tumor Microenvironment , Tumor-Associated Macrophages/metabolism , Aged , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , CD3 Complex/metabolism , Carcinoma/drug therapy , Carcinoma/genetics , Carcinoma/immunology , Cells, Cultured , Chemokine CXCL10/genetics , Drug Resistance, Neoplasm , Female , Humans , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Middle Aged , Neoplasms, Cystic, Mucinous, and Serous/drug therapy , Neoplasms, Cystic, Mucinous, and Serous/genetics , Neoplasms, Cystic, Mucinous, and Serous/immunology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Phenotype , Prognosis , Receptors, Cell Surface/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Tumor-Associated Macrophages/immunology
16.
Bull Exp Biol Med ; 171(3): 367-369, 2021 Jul.
Article En | MEDLINE | ID: mdl-34302206

The phenomenon of multiple primary malignant tumors (MPMT) is characterized by the presence of several primary neoplasms in the same patient. An experimental model of MPMT with one dominating tumor was developed. Female BALB/c nude mice received simultaneous subcutaneous inoculation of Guerin's carcinoma (5×105 tumor cells in 0.5 ml saline) and B16/F10 melanoma (0.5 ml suspension diluted 1:20 with saline). Control females received transplantation of either melanoma or carcinoma alone in the same doses and volumes. In animals with MPMT model, tumors appeared 3-fold faster than after isolated transplantation of melanoma or Guerin's carcinoma and were larger by 7.5 and 2.2 times, respectively; the survival of mice with MPMT was lower. Guerin's carcinoma in the MPMT model metastasized to melanoma and almost completely suppressed its growth. Thus, a MPMT model was created with carcinoma suppressing the malignant growth of melanoma.


Carcinoma/pathology , Melanoma, Experimental/pathology , Neoplasms, Multiple Primary/pathology , Primary Immunodeficiency Diseases/pathology , Urologic Neoplasms/pathology , Animals , Carcinoma/immunology , Cell Line, Tumor , Disease Models, Animal , Female , Injections, Subcutaneous , Melanoma, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Neoplasms, Multiple Primary/immunology , Primary Immunodeficiency Diseases/immunology , Rats , Time Factors , Tumor Burden , Urologic Neoplasms/immunology
17.
Int J Mol Sci ; 22(14)2021 Jul 09.
Article En | MEDLINE | ID: mdl-34299016

Metaplastic breast carcinoma (MBC) is a heterogeneous group of infrequent triple negative (TN) invasive carcinomas with poor prognosis. MBCs have a different clinical behavior from other types of triple negative breast cancer (TNBC), being more resistant to standard chemotherapy. MBCs are an example of tumors with activation of epithelial-mesenchymal transition (EMT). The mechanisms involved in EMT could be responsible for the increase in the infiltrative and metastatic capacity of MBCs and resistance to treatments. In addition, a relationship between EMT and the immune response has been seen in these tumors. In this sense, MBC differ from other TN tumors showing a lower number of tumor-infiltrating lymphocytes (TILS) and a higher percentage of tumor cells expressing programmed death-ligand 1 (PD-L1). A better understanding of the relationship between the immune system and EMT could provide new therapeutic approaches in MBC.


B7-H1 Antigen/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Carcinoma/immunology , Carcinoma/metabolism , Epithelial-Mesenchymal Transition/immunology , B7-H1 Antigen/immunology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cadherins/metabolism , Carcinoma/drug therapy , Carcinoma/genetics , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Lymphocytes, Tumor-Infiltrating/immunology , MicroRNAs/genetics , MicroRNAs/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
18.
J Obstet Gynaecol Res ; 47(9): 2978-2989, 2021 Sep.
Article En | MEDLINE | ID: mdl-34184357

AIMS: Tissue factor pathway inhibitor (TFPI)-2 has recently emerged as a serodiagnostic marker for patients with epithelial ovarian cancer (EOC), especially clear cell carcinoma (CCC). This review discusses the biological properties of TFPI-2 and why serum levels are elevated in CCC patients. METHODS: A comprehensive literature search was conducted in PubMed up until March, 2021. RESULTS: TFPI-2 is a Kunitz-type protease inhibitor and negatively regulates the enzymatic activities, such as plasmin. TFPI-2 has been characterized as a tumor suppressor gene and was frequently downregulated through promoter hypermethylation in various human cancers. In contrast, TFPI-2 was overexpressed only in CCC. TFPI-2 may be involved in the pathophysiology of CCC, possibly through regulation of coagulation system, stabilization of extracellular matrix (ECM), and induction of intracellular signal transduction. TFPI-2 suppresses tissue factor-induced hypercoagulation in a hypoxic environment. TFPI-2, secreted by CCC cells, platelets, and adjacent vascular endothelial cells, may suppress tumor growth and invasion through ECM remodeling. Nuclear TFPI-2 may suppress matrix metalloproteinase production via transcription factors and modulate caspase-mediated cell apoptosis. CCC cells may upregulate the TFPI-2 expression to adapt to survival in the demanding environment. TFPI-2 is secreted by CCC cells and enters the systemic circulation, resulting in elevated blood levels. DISCUSSION: Serum TFPI-2 reflects the overexpression of TFPI-2 in CCC tissues and is a potential serodiagnostic marker. Further research is needed to explore the expression, clinical significance, biological function, and potential mechanism of TFPI-2 in CCC.


Carcinoma , Endothelial Cells , Glycoproteins , Ovarian Neoplasms , Carcinoma/diagnosis , Carcinoma/immunology , Female , Glycoproteins/genetics , Glycoproteins/immunology , Humans , Lipoproteins , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/immunology , Ovary , Serologic Tests
19.
Gynecol Oncol ; 162(2): 421-430, 2021 08.
Article En | MEDLINE | ID: mdl-34088514

OBJECTIVE: To define the pre-treatment tumour immune landscape of clear cell carcinoma of the ovary (CCOC). METHODS: We investigated the infiltration profiles of selected immune cell populations and immune checkpoint proteins that have been previously shown to have prognostic relevance in high grade serous carcinoma of the ovary to determine their association with clinical outcomes in CCOC patients. Using multiplex immunohistochemistry, we evaluated the density of CD3+, FoxP3+, CD8+ T cells, CD20+ B cells and expression of PD-1, PD-L1 and IDO1 immune checkpoints in a cohort of 162 CCOC tumour specimens on a tissue microarray. RESULTS: Increased infiltration of CD3+ CD8- (helper T) cells, CD8+ (cytotoxic T) cells, and CD68+ macrophages significantly associated with shorter disease-free survival, recurrence-free survival and overall survival. Importantly, higher expression of PD-L1 and IDO-1 immune checkpoints was associated with better clinical outcomes. CONCLUSION: Findings from our study are foundational towards the development of immune classifiers and biomarkers of response to immune checkpoint blockade therapy in CCOC.


Carcinoma/mortality , Neoplasm Recurrence, Local/epidemiology , Ovarian Neoplasms/mortality , Ovary/immunology , Tumor Microenvironment/immunology , Adult , Aged , B7-H1 Antigen/analysis , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Carcinoma/drug therapy , Carcinoma/immunology , Disease-Free Survival , Female , Gene Expression Profiling , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Indoleamine-Pyrrole 2,3,-Dioxygenase/analysis , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Middle Aged , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/prevention & control , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/immunology , Ovary/pathology , Prognosis , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tissue Array Analysis , Tumor Microenvironment/drug effects , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/immunology
20.
Eur J Cancer ; 151: 35-48, 2021 07.
Article En | MEDLINE | ID: mdl-33962359

INTRODUCTION: Platinum-based combination chemotherapy is the standard treatment for patients with chemotherapy-eligible metastatic urothelial carcinoma (mUC). Immune-checkpoint inhibitors (ICIs) are currently assessed in this setting. This review aimed to assess the role of ICIs alone or in combination as first-line treatment in chemotherapy-eligible patients with mUC. METHODS: Multiple databases were searched for articles published until November 2020. Studies were deemed eligible if they compared overall survival (OS), progression-free survival (PFS), objective response rates (ORRs), complete response rates (CRRs), durations of response (DORs) and adverse events (AEs) in chemotherapy-eligible patients with mUC. RESULTS: Three studies met our eligibility criteria. ICI combination therapy was associated with significantly better OS and PFS, higher CRR and longer DOR than chemotherapy alone (hazard ratio [HR]: 0.85, 95% confidence interval [CI]: 0.76-0.94, P = 0.002; HR: 0.80, 95% CI: 0.71-0.90, P = 0.0002; odds ratio [OR]: 1.48, 95% CI: 1.12-1.96, P = 0.006; and mean difference: 1.39, 95% CI: 0.31-2.46, P = 0.01, respectively). ICI-chemotherapy combination therapy was also associated with significantly better OS and PFS, higher ORR and CRR and longer DOR than chemotherapy alone. Although OS and PFS benefits of ICI combination therapy were larger in patients with high expression of programmed death-ligand 1 (PD-L1), PD-L1 low expression patients also had a benefit; HR for OS (high PD-L1: HR 0.79 versus low PD-L1: HR 0.89) and PFS (high PD-L1: HR 0.74 versus low PD-L1: HR 0.82). ICI monotherapy was not associated with better oncological outcomes but was associated with better safety outcomes than chemotherapy alone. CONCLUSIONS: Our analysis indicates a superior oncologic benefit to first-line ICI combination therapies in patients with chemotherapy-eligible mUC over standard chemotherapy. In contrast, ICI monotherapy was associated with favorable safety outcomes compared with chemotherapy but failed to show its superiority over chemotherapy in oncological benefits. PD-L1 status alone cannot help guide treatment decision-making. However, caution should be exercised in interpreting the conclusions drawn from this study, given that there is the heterogeneity of the population of interest, risk of bias and the nature of the studies evaluated whose data remain immature or unpublished.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Urothelium/drug effects , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , B7-H1 Antigen/antagonists & inhibitors , Carcinoma/immunology , Carcinoma/mortality , Carcinoma/pathology , Clinical Decision-Making , Female , Humans , Immune Checkpoint Inhibitors/adverse effects , Male , Middle Aged , Patient Selection , Progression-Free Survival , Time Factors , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology , Urothelium/immunology , Urothelium/pathology
...