Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunother Cancer ; 10(5)2022 05.
Article in English | MEDLINE | ID: mdl-35606086

ABSTRACT

BACKGROUND: Cancer immunotherapeutic strategies showed unprecedented results in the clinic. However, many patients do not respond to immuno-oncological treatments due to the occurrence of a plethora of immunological obstacles, including tumor intrinsic mechanisms of resistance to cytotoxic T-cell (TC) attack. Thus, a deeper understanding of these mechanisms is needed to develop successful immunotherapies. METHODS: To identify novel genes that protect tumor cells from effective TC-mediated cytotoxicity, we performed a genetic screening in pancreatic cancer cells challenged with tumor-infiltrating lymphocytes and antigen-specific TCs. RESULTS: The screening revealed 108 potential genes that protected tumor cells from TC attack. Among them, salt-inducible kinase 3 (SIK3) was one of the strongest hits identified in the screening. Both genetic and pharmacological inhibitions of SIK3 in tumor cells dramatically increased TC-mediated cytotoxicity in several in vitro coculture models, using different sources of tumor and TCs. Consistently, adoptive TC transfer of TILs led to tumor growth inhibition of SIK3-depleted cancer cells in vivo. Mechanistic analysis revealed that SIK3 rendered tumor cells susceptible to tumor necrosis factor (TNF) secreted by tumor-activated TCs. SIK3 promoted nuclear factor kappa B (NF-κB) nuclear translocation and inhibited caspase-8 and caspase-9 after TNF stimulation. Chromatin accessibility and transcriptome analyses showed that SIK3 knockdown profoundly impaired the expression of prosurvival genes under the TNF-NF-κB axis. TNF stimulation led to SIK3-dependent phosphorylation of the NF-κB upstream regulators inhibitory-κB kinase and NF-kappa-B inhibitor alpha on the one side, and to inhibition of histone deacetylase 4 on the other side, thus sustaining NF-κB activation and nuclear stabilization. A SIK3-dependent gene signature of TNF-mediated NF-κB activation was found in a majority of pancreatic cancers where it correlated with increased cytotoxic TC activity and poor prognosis. CONCLUSION: Our data reveal an abundant molecular mechanism that protects tumor cells from cytotoxic TC attack and demonstrate that pharmacological inhibition of this pathway is feasible.


Subject(s)
NF-kappa B , Tumor Necrosis Factor-alpha , Apoptosis , Humans , NF-kappa B/metabolism , Phosphorylation , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism
2.
Nat Commun ; 12(1): 1119, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33602930

ABSTRACT

Regulatory CD4+ T cells (Treg) prevent tumor clearance by conventional T cells (Tconv) comprising a major obstacle of cancer immune-surveillance. Hitherto, the mechanisms of Treg repertoire formation in human cancers remain largely unclear. Here, we analyze Treg clonal origin in breast cancer patients using T-Cell Receptor and single-cell transcriptome sequencing. While Treg in peripheral blood and breast tumors are clonally distinct, Tconv clones, including tumor-antigen reactive effectors (Teff), are detected in both compartments. Tumor-infiltrating CD4+ cells accumulate into distinct transcriptome clusters, including early activated Tconv, uncommitted Teff, Th1 Teff, suppressive Treg and pro-tumorigenic Treg. Trajectory analysis suggests early activated Tconv differentiation either into Th1 Teff or into suppressive and pro-tumorigenic Treg. Importantly, Tconv, activated Tconv and Treg share highly-expanded clones contributing up to 65% of intratumoral Treg. Here we show that Treg in human breast cancer may considerably stem from antigen-experienced Tconv converting into secondary induced Treg through intratumoral activation.


Subject(s)
Breast Neoplasms/immunology , Breast Neoplasms/pathology , T-Lymphocytes, Regulatory/immunology , Antigens, Neoplasm/metabolism , Breast Neoplasms/blood , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Clone Cells , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lymphocyte Activation/immunology , Neoplasm Staging , Receptors, Antigen, T-Cell/immunology , Single-Cell Analysis , Th1 Cells/immunology , Transcriptome/genetics
3.
Cancer Immunol Res ; 8(9): 1163-1179, 2020 09.
Article in English | MEDLINE | ID: mdl-32665263

ABSTRACT

The success of cancer immunotherapy is limited by resistance to immune checkpoint blockade. We therefore conducted a genetic screen to identify genes that mediated resistance against CTLs in anti-PD-L1 treatment-refractory human tumors. Using PD-L1-positive multiple myeloma cells cocultured with tumor-reactive bone marrow-infiltrating CTL as a model, we identified calcium/calmodulin-dependent protein kinase 1D (CAMK1D) as a key modulator of tumor-intrinsic immune resistance. CAMK1D was coexpressed with PD-L1 in anti-PD-L1/PD-1 treatment-refractory cancer types and correlated with poor prognosis in these tumors. CAMK1D was activated by CTL through Fas-receptor stimulation, which led to CAMK1D binding to and phosphorylating caspase-3, -6, and -7, inhibiting their activation and function. Consistently, CAMK1D mediated immune resistance of murine colorectal cancer cells in vivo The pharmacologic inhibition of CAMK1D, on the other hand, restored the sensitivity toward Fas-ligand treatment in multiple myeloma and uveal melanoma cells in vitro Thus, rapid inhibition of the terminal apoptotic cascade by CAMK1D expressed in anti-PD-L1-refractory tumors via T-cell recognition may have contributed to tumor immune resistance.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 1/immunology , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/transplantation , Animals , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/immunology , Calcium-Calmodulin-Dependent Protein Kinase Type 1/biosynthesis , Drug Resistance, Neoplasm , Humans , Mice , Multiple Myeloma/immunology , Multiple Myeloma/therapy
4.
EMBO Mol Med ; 7(4): 450-63, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25691366

ABSTRACT

The success of T cell-based cancer immunotherapy is limited by tumor's resistance against killing by cytotoxic T lymphocytes (CTLs). Tumor-immune resistance is mediated by cell surface ligands that engage immune-inhibitory receptors on T cells. These ligands represent potent targets for therapeutic inhibition. So far, only few immune-suppressive ligands have been identified. We here describe a rapid high-throughput siRNA-based screening approach that allows a comprehensive identification of ligands on human cancer cells that inhibit CTL-mediated tumor cell killing. We exemplarily demonstrate that CCR9, which is expressed in many cancers, exerts strong immune-regulatory effects on T cell responses in multiple tumors. Unlike PDL1, which inhibits TCR signaling, CCR9 regulates STAT signaling in T cells, resulting in reduced T-helper-1 cytokine secretion and reduced cytotoxic capacity. Moreover, inhibition of CCR9 expression on tumor cells facilitated immunotherapy of human tumors by tumor-specific T cells in vivo. Taken together, this method allows a rapid and comprehensive determination of immune-modulatory genes in human tumors which, as an entity, represent the 'immune modulatome' of cancer.


Subject(s)
CD8-Positive T-Lymphocytes , Immunity, Cellular , Immunotherapy/methods , Neoplasms, Experimental , RNA Interference , Th1 Cells , Animals , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Female , Humans , MCF-7 Cells , Mice , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Receptors, CCR/immunology , Th1 Cells/immunology , Th1 Cells/pathology
5.
J Immunol ; 190(5): 2464-71, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23359505

ABSTRACT

The antitumor effects of paclitaxel are generally attributed to the suppression of microtubule dynamics resulting in defects in cell division. New data demonstrated that in ultralow noncytotoxic concentrations, paclitaxel modulated in immune cells in vitro the activity of small Rho GTPases, the key regulators of intracellular actin dynamics. However, the immunomodulatory properties of paclitaxel in vivo have not been evaluated. In this study, using the ret transgenic murine melanoma model, which mimics human cutaneous melanoma, we tested effects of ultralow noncytotoxic dose paclitaxel on functions of myeloid-derived suppressor cells (MDSCs), chronic inflammatory mediators, and T cell activities in the tumor microenvironment in vivo. Administration of paclitaxel significantly decreased accumulation and immunosuppressive activities of tumor-infiltrating MDSCs without alterations of the bone marrow hematopoiesis. This was associated with the inhibition of p38 MAPK activity, TNF-α and production, and S100A9 expression in MDSCs. The production of mediators of chronic inflammation in the tumor milieu also was diminished. Importantly, reduced tumor burden and increased animal survival upon paclitaxel application was mediated by the restoration of CD8 T cell effector functions. We suggest that the ability of paclitaxel in a noncytotoxic dose to block the immunosuppressive potential of MDSCs in vivo represents a new therapeutic strategy to downregulate immunosuppression and chronic inflammation in the tumor microenvironment for enhancing the efficacy of concomitant anticancer therapies.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Inflammation/drug therapy , Melanoma/drug therapy , Myeloid Cells/drug effects , Paclitaxel/therapeutic use , Skin Neoplasms/drug therapy , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Calgranulin B/genetics , Calgranulin B/immunology , Chronic Disease , Dose-Response Relationship, Immunologic , Humans , Immunosuppression Therapy , Inflammation/complications , Inflammation/immunology , Inflammation/pathology , Melanoma/complications , Melanoma/immunology , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/immunology , Myeloid Cells/immunology , Myeloid Cells/pathology , Paclitaxel/pharmacology , Primary Cell Culture , Skin Neoplasms/complications , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Spleen/cytology , Spleen/drug effects , Spleen/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology
6.
J Invest Dermatol ; 133(6): 1610-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23223128

ABSTRACT

Low-dose cyclophosphamide (CP) therapy induces immunogenic tumor cell death and decreases regulatory T cell (Treg) numbers in mice with transplantable tumors. Using the ret transgenic murine melanoma model that resembles human melanoma, we detected no beneficial antitumor effects with such treatment, despite a decrease in Tregs. On the contrary, low-dose CP enhanced the production of chronic inflammatory mediators in melanoma lesions associated with increased accumulation of Gr1(+)CD11b(+) myeloid-derived suppressor cells (MDSCs), which exhibit elevated suppressive activity and nitric oxide (NO) production as well as inhibition of T-cell proliferation. Moreover, the frequencies of CD8(+) T cells in the tumors and their ability to produce perforin were decreased. To study whether the observed CP-induced MDSC expansion and activation also occurs under chronic inflammatory tumor-free conditions, mice exhibiting chronic inflammation were treated with CP. Similar to tumor-bearing mice, CP-treated inflamed mice displayed elevated levels of MDSCs with enhanced production of NO, reactive oxygen species, and a suppressed in vivo natural killer (NK) cell cytotoxic activity indicating CP effects on the host immune system independent of the tumor. We suggest that melanoma therapy with low-dose CP could be efficient only when combined with the neutralization of MDSC immunosuppressive function and chronic inflammatory microenvironment.


Subject(s)
Cyclophosphamide/pharmacology , Immunosuppression Therapy/methods , Melanoma/drug therapy , Skin Neoplasms/drug therapy , T-Lymphocytes, Regulatory/drug effects , Animals , Chronic Disease , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Immunosuppressive Agents/pharmacology , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Melanoma/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/cytology , Myeloid Cells/drug effects , Myeloid Cells/immunology , Proto-Oncogene Proteins c-ret/genetics , Skin Neoplasms/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
7.
Org Lett ; 14(20): 5218-21, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-23025410

ABSTRACT

Aspartimide formation is one of the major obstacles that impedes the solid phase synthesis of large peptides and proteins. Until now, no cost-effective strategy to suppress this side reaction has been developed. Here it is demonstrated that addition of small amounts of organic acids to the standard Fmoc cleavage agent piperidine efficiently prevents formation of aspartimide side products. This effect is shown to be virtually independent of the acid strength.


Subject(s)
Acids/chemistry , Aspartic Acid/analogs & derivatives , Aspartic Acid/chemical synthesis , Hydrogen-Ion Concentration , Molecular Structure , Solid-Phase Synthesis Techniques
8.
J Immunotoxicol ; 9(3): 292-300, 2012.
Article in English | MEDLINE | ID: mdl-22283566

ABSTRACT

Myeloid cells play a key role in the outcome of anti-tumor immunity and response to anti-cancer therapy, since in the tumor microenvironment they may exert both stimulatory and inhibitory pressures on the proliferative, angiogenic, metastatic, and immunomodulating potential of tumor cells. Therefore, understanding the mechanisms of myeloid regulatory cell differentiation is critical for developing strategies for the therapeutic reversal of myeloid derived suppressor cell (MDSC) accumulation in the tumor-bearing hosts. Here, using an in vitro model system, several potential mechanisms of the direct effect of paclitaxel on MDSC were tested, which might be responsible for the anti-tumor potential of low-dose paclitaxel therapy in mice. It was hypothesized that a decreased level of MDSC in vivo after paclitaxel administration might be due to (i) the blockage of MDSC generation, (ii) an induction of MDSC apoptosis, or (iii) the stimulation of MDSC differentiation. The results revealed that paclitaxel in ultra-low concentrations neither increased MDSC apoptosis nor blocked MDSC generation, but stimulated MDSC differentiation towards dendritic cells. This effect of paclitaxel was TLR4-independent since it was not diminished in cell cultures originated from TLR4-/- mice. These results support a new concept that certain chemotherapeutic agents in ultra-low non-cytotoxic doses may suppress tumor progression by targeting several cell populations in the tumor microenvironment, including MDSC.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Differentiation/drug effects , Dendritic Cells/immunology , Myeloid Cells/immunology , Neoplasms/immunology , Paclitaxel/pharmacology , Tumor Microenvironment/immunology , Animals , Cell Line, Tumor , Male , Mice , Neoplasms/therapy , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...