Project description:We compared gene expression profile between healthy-donor peripheral monocytes and glioblastoma-patient peripheral monocytes as well as glioblastoma-patient peripheral monocytes with matched tumor-infiltrating myeloid cells.
Project description:MicroRNAs (miRNAs) are non-coding molecules involved in post-transcriptional gene regulation that have been shown to modulate tumor cell proliferation and apoptosis and to act as oncogenes or tumor-suppressor genes. Although miRNAs have been linked to tumor progression, the connection between tumor-mediated immune modulation and miRNAs has yet to be explored. Specifically, how the miRNA dysregulation affects the monocyte-derived glioblastoma-infiltrating macrophages, the most abundant immune cell population within the glioblastoma microenvironment, and their immune suppressive properties has not been evaluated to date. Here we managed to purify the glioblastoma-infiltrating macrophages from the tumor microenvironment and compared their miRNA expression profile with the matched peripheral monocytes from the peripheral blood of the same GBM patient as well as with healthy donors. Of note, several most down-regulated miRNA candidates revealed in this study, including miR-142-3p, were also known for their role in mediating tumor-associated immunosuppression. These results suggest a novel approach to identify miRNA immune therapeutics using a two-step process: 1) screen miRNA expression from tumor-associated immune cells relative to normal immune cell, and 2) select and prioritize potential candidates on the basis of binding to immunosuppressive pathways or mechanisms. In the study presented here, 12 samples, including peripheral monocyte samples from 4 healthy donors, peripheral monocytes from 4 GBM patients and matched tumor-infiltrating macrophages extracted from the glioblastoma microenvironment, were used to acquire the miRNA expression profiles of 1732 unique mature miRNA sequences via the Phalanx Human miRNA OneArray Microarray v3 Platform.
Project description:MicroRNAs (miRNAs) are non-coding molecules involved in post-transcriptional gene regulation that have been shown to modulate tumor cell proliferation and apoptosis and to act as oncogenes or tumor-suppressor genes. Although miRNAs have been linked to tumor progression, the connection between tumor-mediated immune modulation and miRNAs has yet to be explored. Specifically, how the miRNA dysregulation affects the monocyte-derived glioblastoma-infiltrating macrophages, the most abundant immune cell population within the glioblastoma microenvironment, and their immune suppressive properties has not been evaluated to date. Here we managed to purify the glioblastoma-infiltrating macrophages from the tumor microenvironment and compared their miRNA expression profile with the matched peripheral monocytes from the peripheral blood of the same GBM patient as well as with healthy donors. Of note, several most down-regulated miRNA candidates revealed in this study, including miR-142-3p, were also known for their role in mediating tumor-associated immunosuppression. These results suggest a novel approach to identify miRNA immune therapeutics using a two-step process: 1) screen miRNA expression from tumor-associated immune cells relative to normal immune cell, and 2) select and prioritize potential candidates on the basis of binding to immunosuppressive pathways or mechanisms.
Project description:CD4+ helper T (Th) cells are critical regulators of immune responses but their role in breast cancer is currently unknown. This work aims to characterize Th cells infiltrating invasive primary human breast tumors, analyze the influence by the tumor microenvironment and identify Th cell specific prognostic gene signatures. CD4+ T cells isolated from the tumor (TIL), axillary lymph node (LN) and blood (PB) of 10 patients were analyzed on Affymetrix U133 Plus 2.0 arrays. A confirmation set of 60 patients were studied by flow cytometry, qRT-PCR or immunohistochemistry and analyzed according to the extent of the tumor immune infiltrate. Gene expression profiles of freshly isolated TIL were also compared with TIL that had been rested overnight or with CD4+ T cells [non-stimulated (NS) or stimulated (S)] from healthy donor PB treated with tumor supernatant (SN). Analysis of CD4+ TIL by comparing their expression profiles to those of their conterparts from patient axillary lymph nodes and peripheral blood and healthy donor blood CD4+ T cells were isolated from primary tumors, axillary lymph nodes and peripheral blood of 10 patients with invasive breast carcinomas and blood of 4 healthy donors and analyzed on Affymetrix U133 Plus 2.0 arrays
Project description:<p>PD-1 is an important immune checkpoint inhibitor that shows great promise in the clinic, particularly for melanoma and lung cancers. Since PD-1 is also expressed on infiltrating CD4+ Treg and Teffector cells in glioblastoma, we sought to better understand the role of PD-1 on these infiltrating CD4+ Treg and Teffector cells. To this end, we performed functional and transcriptional profiling using CD4+ Treg and Teffector cells isolated from healthy donors and glioblastoma patients (from both tumors and blood).</p>
Project description:It was previously demonstrated that myeloid cells assume a distinct transcriptional profile when infiltrating the brain during chronic inflammation. We demonstrated that neutrophils infiltrate the brain during pancreatic cancer. To determine if the transcriptional profile of brain-infiltrating neutrophils is distinct from those infiltrating other organs, we compared the gene signatures of brain-infiltrating neutrophils, circulating neutrophils, liver-infiltrating neutrophils, and tumor-infiltrating neutrophils to that of circulating neutrophils from healthy mice.
Project description:This pilot clinical trial studies low-dose total body irradiation and donor peripheral blood stem cell transplant followed by donor lymphocyte infusion in treatment patients with non-Hodgkin lymphoma, chronic lymphocytic leukemia, or multiple myeloma. Giving total-body irradiation before a donor peripheral blood stem cell transplant helps stop the growth of cells in the bone marrow, including normal blood-forming cells (stem cells) and cancer cells. When healthy stem cells from a donor are infused into the patient they may help the patient’s bone marrow make stem cells, red blood cells, white blood cells, and platelets. Once the donated stem cells begin working, the patient’s immune system may see the remaining cancer cells as not belonging in the patient’s body and destroy them. Giving an infusion of the donor’s white blood cells (donor lymphocyte infusion) may boost this effect.
Project description:Peripheral blood monocytes are the starting material utilized in conventional dendritic cell (DC) vaccination for the treatment of a broad range of malignancies. While the use of cytokines and growth factors to polarize monocyte-derived DC to distinct phenotypes is well-established, little is known about the contributions of distinct human monocyte subsets to monocyte-derived DC function and patient responses to vaccination. To investigate the status of monocyte subsets in cancer patients and following culture into DC, we isolated classical (C-Mo), intermediate (I-Mo), and non-classical (NC-Mo) from the peripheral blood of renal cell carcinoma (RCC) patients prior to DC vaccination (NCT00085436) and from anonymous healthy donors. Patients treated with DC vaccination who were long term survivors (>100 months survival) had a unique monocyte signature with a two-fold higher percentage of NC-Mo in pretreatment peripheral blood compared to other RCC patients. RCC patient monocytes from each subset were transcriptionally distinct from healthy donor monocytes. Further transcriptional analysis determined that each monocyte subset was characterized by a discrete gene expression profile before and after DC maturation. Phenotypic analysis showed that DC derived from NC-Mo expressed higher levels of CD80, CD83, CD86, HLA-DR, and CD40 compared to DC originating from C-Mo and secreted increased amounts of IL-12p70 following CD40L stimulation. Collectively, these findings establish that DC derived from NC-Mo are potent antigen presenting cells and provide the foundation for future vaccination strategies that enrich NC-Mo prior to DC maturation.
Project description:Peripheral blood monocytes are the starting material utilized in conventional dendritic cell (DC) vaccination for the treatment of a broad range of malignancies. While the use of cytokines and growth factors to polarize monocyte-derived DC to distinct phenotypes is well-established, little is known about the contributions of distinct human monocyte subsets to monocyte-derived DC function and patient responses to vaccination. To investigate the status of monocyte subsets in cancer patients and following culture into DC, we isolated classical (C-Mo), intermediate (I-Mo), and non-classical (NC-Mo) from the peripheral blood of renal cell carcinoma (RCC) patients prior to DC vaccination (NCT00085436) and from anonymous healthy donors. Patients treated with DC vaccination who were long term survivors (>100 months survival) had a unique monocyte signature with a two-fold higher percentage of NC-Mo in pretreatment peripheral blood compared to other RCC patients. RCC patient monocytes from each subset were transcriptionally distinct from healthy donor monocytes. Further transcriptional analysis determined that each monocyte subset was characterized by a discrete gene expression profile before and after DC maturation. Phenotypic analysis showed that DC derived from NC-Mo expressed higher levels of CD80, CD83, CD86, HLA-DR, and CD40 compared to DC originating from C-Mo and secreted increased amounts of IL-12p70 following CD40L stimulation. Collectively, these findings establish that DC derived from NC-Mo are potent antigen presenting cells and provide the foundation for future vaccination strategies that enrich NC-Mo prior to DC maturation.