scholarly journals Cellular and gene signatures of tumor-infiltrating dendritic cells and natural-killer cells predict prognosis of neuroblastoma

2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Ombretta Melaiu ◽  
Marco Chierici ◽  
Valeria Lucarini ◽  
Giuseppe Jurman ◽  
Libenzio Adrian Conti ◽  
...  

AbstractTumor-infiltrating lymphocytes play an essential role in improving clinical outcome of neuroblastoma (NB) patients, but their relationship with other tumor-infiltrating immune cells in the T cell-inflamed tumors remains poorly investigated. Here we show that dendritic cells (DCs) and natural killer (NK) cells are positively correlated with T-cell infiltration in human NB, both at transcriptional and protein levels, and associate with a favorable prognosis. Multiplex imaging displays DC/NK/T cell conjugates in the tumor microenvironment of low-risk NB. Remarkably, this connection is further strengthened by the identification of gene signatures related to DCs and NK cells able to predict survival of NB patients and strongly correlate with the expression of PD-1 and PD-L1. In summary, our findings unveil a key prognostic role of DCs and NK cells and indicate their related gene signatures as promising tools for the identification of clinical biomarkers to better define risk stratification and survival of NB patients.

2021 ◽  
Vol 49 (1) ◽  
pp. 030006052098153
Author(s):  
Qing Bi ◽  
Yang Liu ◽  
Tao Yuan ◽  
Huizhen Wang ◽  
Bin Li ◽  
...  

Objective The role of tumor-infiltrating lymphocytes (TILs) has not yet been characterized in sarcomas. The aim of this bioinformatics study was to explore the effect of TILs on sarcoma survival and genome alterations. Methods Whole-exome sequencing, transcriptome sequencing, and survival data of sarcoma were obtained from The Cancer Genome Atlas. Immune infiltration scores were calculated using the Tumor Immune Estimation Resource. Potential associations between abundance of infiltrating TILs and survival or genome alterations were examined. Results Levels of CD4+ T cell infiltration were associated with overall survival of patients with pan-sarcomas, and higher CD4+ T cell infiltration levels were associated with better survival. Somatic copy number alterations, rather than mutations, were found to correlate with CD4+ T cell infiltration levels. Conclusions This data mining study indicated that CD4+ T cell infiltration levels predicted from RNA sequencing could predict sarcoma prognosis, and higher levels of CD4+ T cells infiltration indicated a better chance of survival.


2014 ◽  
Vol 2014 ◽  
pp. 1-9 ◽  
Author(s):  
Xiaoling Qian ◽  
Xian Wang ◽  
Hongchuan Jin

Cell transfer therapy for cancer has made a rapid progress recently and the immunotherapy has been recognized as the fourth anticancer modality after operation, chemotherapy, and radiotherapy. Lymphocytes used for cell transfer therapy include dendritic cells, natural killer (NK) cells, and T lymphocytes such as tumor-infiltrating lymphocytes (TILs) and cytotoxic T lymphocytes (CTLs). In vitro activated or engineered immune cells can traffic to cancer tissues to elicit persistent antitumor immune response which is very important especially after immunosuppressive treatments such as chemotherapy. In this review, we overviewed recent advances in the exploration of dendritic cells, NK cells, and T cells for the treatment of human cancer cells.


1990 ◽  
Vol 72 (4) ◽  
pp. 616-618 ◽  
Author(s):  
Jesús Vaquero ◽  
Santiago Coca ◽  
Rosa Magallón ◽  
Patricia Pontón ◽  
Roberto Martinez

✓ A monoclonal antibody against the surface marker IOT-10 of natural killer (NK) cells was used to investigate the presence and distribution of these cells in a series of nine primary intracranial germinomas. In all of these tumors, IOT-10-positive NK cells were found in small numbers, mainly distributed among the tumor cells. The data obtained in the present study suggest that the presence of NK cells in primary intracranial germinomas can be influenced by factors other than the mere quantity of tumor-infiltrating lymphocytes.


2019 ◽  
Vol 98 (13) ◽  
pp. 1480-1487 ◽  
Author(s):  
Y. Xiao ◽  
H. Li ◽  
L. Mao ◽  
Q.C. Yang ◽  
L.Q. Fu ◽  
...  

T cells and dendritic cells (DCs) that are positive for the tissue-resident marker CD103 play a vital role in antitumor immunity. In this study, multiplexed immunohistochemistry was applied to stain CD103 and the T-cell marker CD8 as well as the DC marker CD11c on formalin-fixed, paraffin-embedded oral squamous cell carcinoma (OSCC) tissues. Then, the density of CD103+CD8+ and CD103+CD11c+ tumor-infiltrating lymphocytes (TILs) in the intratumoral and stromal regions was calculated, and the correlation of CD103+CD8+ TIL and CD103+CD11c+ TIL density with OSCC patient prognosis was analyzed. The results revealed that CD103+CD8+ TILs and CD103+CD11c+ TILs were abundant in the stromal region and that increased stromal CD103+CD8+ TIL and intratumoral CD103+CD11c+ TIL density indicated a favorable prognosis. Moreover, we freshly isolated TILs from OSCC samples and performed flow cytometry to verify that CD103+CD8+ TILs display a tissue-resident memory T-cell (Trm) phenotype, and we discriminated CD103+CD11c+ TILs from tumor-associated macrophages.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A98-A98
Author(s):  
Garima Kaushik ◽  
Bhavna Verma ◽  
Amy Wesa

BackgroundThe preclinical screening of immune-modulatory therapies suffers from the absence of models that recapitulate in vivo heterogeneous tumor microenvironment (TME). 3D tumor organoid cultures provide a model that closely mimics in situ tumor architecture and is being aggressively used to evaluate therapeutic efficacy ex vivo. A vastly heterogenous TME impacts patient treatment response, and there is a dearth of human tumor models (2D or 3D), that mimic in vivo diversity of TME, including infiltrating immune populations. 3D organoid cultures typically contain neoplastic epithelium; however, they fall short in representing tumor to tumor-infiltrating lymphocytes (TILs) interactions, limiting their ability to generate a clinically relevant response to immunotherapeutics. Addition of immune cells from unrelated donors to organoids can simulate that microenvironment but is complicated by T cell alloreactivity. Here we describe 3D patient-derived xenograft organoid (PDXO) co-cultures with matching autologous human TILs to recapitulate the tumor-specific immune response, leveraging confocal high content analysis and luminex multiplex assays. This platform allows the evaluation and high throughput screening of novel immune targeting agents to determine impacts on patient-derived T cell function, T cell infiltration, and tumor cytotoxicity.MethodsSurgical resections from patients were used to generate patient-derived xenografts and tumor-infiltrating lymphocytes in parallel. PDX were resected and digested to establish PDXO. TILs and organoids from the same patient were fluorescent labeled and cultured together for four days to evaluate tumor infiltration and drug cytotoxicity in 3D cultures. CellInsight CX7 high content imaging platform was used to trace TILs and cancer cells and evaluate T cell infiltration and tumor cell killing in the presence and absence of immuno-modulatory therapies.ResultsPDXO were established to mimic in vivo tumor biology. Tumor-specific TILs were successfully expanded and characterized by flow cytometry. Co-culture resulted in TIL infiltration in organoids from day one in culture and increased over four days. Cytotoxicity and TIL infiltration were quantified using fluorescent dyes via high throughput imaging platform. Significantly enhanced TIL infiltration was observed in autologous co-cultures compared to non-autologous co-cultures. The established unique autologous PDXO immune organoid co-cultures could be used as an improved simulation of the modulatory activity of therapeutic agents in patient-specific T cells against their own tumors.ConclusionsPatient autologous TILs – PDXO co-culture platform is an advanced model for evaluating IO therapeutics with the tumor-specific immune microenvironment. The platform provides an opportunity for precision medicine and high throughput drug screening of immuno-modulatory therapies.Ethics ApprovalThe study was approved by Champions Oncology’s Institutional Animal Care and Use Committee (IACUC).


Blood ◽  
2008 ◽  
Vol 112 (5) ◽  
pp. 1776-1783 ◽  
Author(s):  
Sophie Agaugué ◽  
Emanuela Marcenaro ◽  
Bruna Ferranti ◽  
Lorenzo Moretta ◽  
Alessandro Moretta

Abstract Dendritic cells (DCs) play a crucial role in naive T-cell priming. Recent data suggested that natural killer (NK) cells can influence the capability of DCs to promote Th1 polarization. This regulatory function is primarily mediated by cytokines released in the microenvironment during inflammatory responses involving NK cells. In this study, we show that human NK cells exposed for short time to interleukin (IL)–12, IL-2, or IL-18, promote distinct pathways of Th1 priming. IL-12– or IL-2–conditioned NK cells induce maturation of DCs capable of priming IFN-γ–producing Th1 cells. On the other hand, IL-18–conditioned NK cells induce Th1 polarization only when cocultured with both DCs and T cells. In this case, IL-2 released by T cells and IL-12 derived from DCs during the priming process promote interferon (IFN)–γ production. In contrast, when NK cells are exposed to IL-4, nonpolarized T cells releasing only low levels of IL-2 are generated. Thus, the prevalence of IL-12, IL-2, IL-18, or IL-4 at inflammatory sites may differentially modulate the NK-cell interaction with DCs, leading to different outcomes in naive T-cell polarization.


2020 ◽  
Vol 105 (7) ◽  
pp. 2217-2228 ◽  
Author(s):  
Wu-Hu Zhang ◽  
Wen-Quan Wang ◽  
He-Li Gao ◽  
Shuai-Shuai Xu ◽  
Shuo Li ◽  
...  

Abstract Objective This study retrospectively characterized the immune infiltrating profile in nonfunctional pancreatic neuroendocrine tumors (NF-PanNETs). Methods Tumor tissues from the 109-patient Fudan cohort and a 73-patient external validation set were evaluated by immunohistochemistry for 9 immune cell types: tumor-infiltrating neutrophils (TINs), tumor-associated macrophages (TAMs), CD11c+ dendritic cells, anti-NCR1+ natural killer (NK) cells, CD4+ and CD8+ T cells, CD45RO+ memory T cells, FOXP3+ regulatory T cells (Tregs), and CD20+ B cells. Results TINs were primarily distributed in the intratumoral area, dendritic cells and NK cells were scattered evenly in intratumoral and stromal areas, and Tregs were rarely detected. The remaining 5 cell types were primarily present in peritumoral stroma. Total TINs (P < .001) and TAMs (P = .002) increased as NF-PanNET grade rose. Kaplan-Meier analyses showed that high intratumoral TINs, total TAMs, and stromal CD4+ T-cell infiltration correlated with shorter recurrence-free survival (RFS, P = .010, P = .027, and P = .035, respectively) and overall survival (OS, P = .017, P = .029, and P = .045, respectively). Additionally, high intratumoral CD8+ T cell infiltration correlated with prolonged RFS (P = .039). Multivariate Cox regression demonstrated that intratumoral TINs, World Health Organization (WHO) classification, and eighth edition of the American Joint Committee on Cancer tumor-node-metastasis staging system (AJCC8th TNM) were independent factors for RFS (P = .043, P = .023, and P = .029, respectively), whereas intratumoral TINs and WHO classification were independent factors for OS (P = .010 and P = .007, respectively). Furthermore, the combination of TINs, WHO classification, and AJCC8th TNM remarkably improved prognostic accuracy for RFS. These results have been verified in the external validation set. Conclusion Intratumoral TINs are an independent and unfavorable predictor of postoperative NF-PanNETs. A combination of TINs, WHO classification, and AJCC8th TNM could improve prognostic accuracy for RFS.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4102-4102
Author(s):  
Leen Willems ◽  
Omer Rutgeerts ◽  
Caroline Lenaerts ◽  
Stefaan Van Gool ◽  
Mark Waer ◽  
...  

Abstract Abstract 4102 Donor leucocyte infusion (DLI) after alloHSCT can induce strong graft-versus-leukemia (GvL) effects, inspiring investigators to examine this approach for solid tumors resistant to conventional therapies (Grivas et al. Curr Clin Pharmacol. 2011). However, DLI produces graft-versus-host disease (GvHD). Recipient-type leucocyte infusion (RLI) is currently being explored clinically as a means to induce GvL without risk of GvHD (clinicaltrials.gov; Rubio et al. Blood 2003; De Somer et al. Haematologica. 2011). High-risk neuroblastoma carries a bleak prognosis despite aggressive treatment with chemo-, radiotherapy and autologous HSCT. Clinical observations in such patients suggest that alloHSCT may produce a graft-vs-neuroblastoma effect (Kanold et al. Bone Marrow Transplantation. 2008). In mice, alloHSCT delays local neuroblastoma growth, and adoptive transfer of tumor-pulsed donor dendritic cells and donor leucocytes enhances this effect (Ash et al. Cancer Immunol Immunother. 2009, Br J Cancer 2010). In this study, we show that not only DLI by itself, but also RLI enhances the local anti-neuroblastoma effect of alloHSCT in mice. MHC-mismatched [C57BL/6 (H-2Kb) → A/J (H-2Kk)] bone marrow chimeras were given a subcutaneous inoculation with 1 × 106 Neuro2A cells (A/J neuroblastoma) on day 14 post HSCT. On day 21, we performed adoptive transfer of 10 × 106 donor splenocytes (DLI), 50 × 106 recipient splencoytes (RLI) and/or 1 × 106 recipient-type MACS-isolated DX5+ NK cells. We measured tumor volume twice weekly using a caliper (volume = width2 × length × 0,4). Validation of this model showed progressive tumor growth and mortality as a result of metastasis. Peripheral blood chimerism and tumor infiltrating lymphocytes were studied using flow cytometry. AlloHSCT chimeras developed mixed donor T cell chimerism by day 21 and full donor chimerism by day 76 post HSCT. DLI induced a conversion to full donor T cell chimerism and RLI induced a complete loss of donor T cells chimerism, both within 1 week. AlloHSCT chimeras showed reduced local growth of subcutaneous neuroblastoma tumors relative to synHSCT chimeras. This delay in tumor growth was enhanced not only by DLI, but also by RLI; DLI provoked lethal GvHD whereas mice treated with RLI remained healthy. Within tumor-infiltrating lymphocytes, T and NK cell chimerism mirrored the systemic chimerism changes seen after RLI and DLI, associated with an increased intratumoral CD8/CD4 ratio, CD8+ T-cell IFN-γ-expression and NK-cell Granzyme B-expression. This indicates a close relation between lymphohematopoietic alloreactivity and the anti-tumor effect, and suggests that the anti-tumor mechanism of DLI and RLI involves not only CD8+ T cells but also cytotoxic NK cells. The baseline antitumor effect seen in alloHSCT chimeras was also accompanied by increased Granzyme B expression by intratumoral NK-cells, supporting a role for NK cells also in this baseline antitumor effect. In vivo Neuro2A-inoculation experiments in (poly(I:C)-treated) C57BL/6, TCR−/− C57BL/6 and A/J mice, and in vitro NK cytotoxicity experiments showed that 1° donor T cells critically resist Neuro2A cells, 2° donor NK cells exhibit spontaneous cytotoxic reactivity to Neuro2A that is enhanced by NK activation, but also that 3° syngeneic NK cells may acquire reactivity to Neuro2A cells provided they are activated. Interestingly, when RLI was given, the intratumoral NK-cell frequency declined markedly, and adoptive transfer of additional NK cells obtained from naïve A/J mice enhanced the local antitumor effect of RLI. This supports the hypothesis that, in addition to donor NK cells, also syngeneic NK cells can play a critical role in mediating a growth-limiting effect on local neuroblastoma. Along this line, we observed that also in synHSCT mice, which showed a reduced local neuroblastoma growth relative to naïve mice, the intratumoral NK cells showed increased FasL-expression. These are the first experimental data showing that RLI after alloHSCT may induce immune-mediated anti-neuroblastoma effects, and that NK cells, in particular syngeneic NK cells, may play a critical role herein. Our data support the exploration of post alloHSCT adoptive cell therapy with recipient-type T and NK cells to produce enhanced immune antitumor effects for high-risk neuroblastoma, and potentially for other solid tumors that are unresponsive to traditional therapies, without the risk of GvHD. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document