Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Cancer Immunol Immunother ; 72(11): 3461-3474, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37528320

ABSTRACT

Myeloid-derived suppressor cells (MDSC) have been linked to loss of immune effector cell function through a variety of mechanisms such as the generation of reactive oxygen and nitrogen species and the production of inhibitory cytokines. Our group has shown that signaling through Bruton's tyrosine kinase (BTK) is important for MDSC function. Ibrutinib is an orally administered targeted agent that inhibits BTK activation and is currently used for the treatment of B cell malignancies. Using a syngeneic murine model of melanoma, the effect of BTK inhibition with ibrutinib on the therapeutic response to systemic PD-L1 blockade was studied. BTK was expressed by murine MDSC and their activation was inhibited by ibrutinib. Ibrutinib was not directly cytotoxic to cancer cells in vitro, but it inhibited BTK activation in MDSC and reduced expression of inducible nitric oxide synthase (NOS2) and production of nitric oxide. Ibrutinib treatments decreased the levels of circulating MDSC in vivo and increased the therapeutic efficacy of anti-PD-L1 antibody treatment. Gene expression profiling showed that ibrutinib decreased Cybb (NOX2) signaling, and increased IL-17 signaling (upregulating downstream targets Mmp9, Ptgs2, and S100a8). These results suggest that further exploration of MDSC inhibition could enhance the immunotherapy of advanced melanoma.PrécisInhibition of Bruton's tyrosine kinase, a key enzyme in myeloid cellular function, improves therapeutic response to an anti-PD-L1 antibody in an otherwise fairly resistant murine melanoma model.


Subject(s)
Antineoplastic Agents , Melanoma , Myeloid-Derived Suppressor Cells , Humans , Mice , Animals , Agammaglobulinaemia Tyrosine Kinase/metabolism , Protein-Tyrosine Kinases , Myeloid-Derived Suppressor Cells/metabolism , B7-H1 Antigen , Immunotherapy , Antineoplastic Agents/therapeutic use , Melanoma/drug therapy
2.
Leukemia ; 36(1): 155-164, 2022 01.
Article in English | MEDLINE | ID: mdl-34312462

ABSTRACT

In this phase I/II clinical trial, we investigated the safety and efficacy of high doses of mb-IL21 ex vivo expanded donor-derived NK cells to decrease relapse in 25 patients with myeloid malignancies receiving haploidentical stem-cell transplantation (HSCT). Three doses of donor NK cells (1 × 105-1 × 108 cells/kg/dose) were administered on days -2, +7, and +28. Results were compared with an independent contemporaneously treated case-matched cohort of 160 patients from the CIBMTR database.After a median follow-up of 24 months, the 2-year relapse rate was 4% vs. 38% (p = 0.014), and disease-free survival (DFS) was 66% vs. 44% (p = 0.1) in the cases and controls, respectively. Only one relapse occurred in the study group, in a patient with the high level of donor-specific anti-HLA antibodies (DSA) presented before transplantation. The 2-year relapse and DFS in patients without DSA was 0% vs. 40% and 72% vs. 44%, respectively with HR for DFS in controls of 2.64 (p = 0.029). NK cells in recipient blood were increased at day +30 in a dose-dependent manner compared with historical controls, and had a proliferating, mature, highly cytotoxic, NKG2C+/KIR+ phenotype.Administration of donor-derived expanded NK cells after haploidentical transplantation was safe, associated with NK cell-dominant immune reconstitution early post-transplant, preserved T-cell reconstitution, and improved relapse and DFS. TRIAL REGISTRATION: NCT01904136 ( https://clinicaltrials.gov/ct2/show/NCT01904136 ).


Subject(s)
Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/mortality , Killer Cells, Natural/transplantation , Neoplasm Recurrence, Local/therapy , Unrelated Donors/statistics & numerical data , Adolescent , Adult , Aged , Case-Control Studies , Female , Follow-Up Studies , Hematologic Neoplasms/immunology , Hematologic Neoplasms/pathology , Humans , Killer Cells, Natural/immunology , Male , Middle Aged , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Prognosis , Survival Rate , Transplantation Conditioning , Transplantation, Haploidentical , Young Adult
3.
Blood Adv ; 5(22): 4605-4618, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34559190

ABSTRACT

The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that regulates cellular processes in cancer and immunity, including innate immune cell development and effector function. However, the transcriptional repertoire through which AHR mediates these effects remains largely unexplored. To elucidate the transcriptional elements directly regulated by AHR in natural killer (NK) cells, we performed RNA and chromatin immunoprecipitation sequencing on NK cells exposed to AHR agonist or antagonist. We show that mature peripheral blood NK cells lack AHR, but its expression is induced by Stat3 during interleukin-21-driven activation and proliferation, coincident with increased NCAM1 (CD56) expression resulting in a CD56bright phenotype. Compared with control conditions, NK cells expanded in the presence of the AHR antagonist, StemRegenin-1, were unaffected in proliferation or cytotoxicity, had no increase in NCAM1 transcription, and maintained the CD56dim phenotype. However, it showed altered expression of 1004 genes including those strongly associated with signaling pathways. In contrast, NK cells expanded in the presence of the AHR agonist, kynurenine, showed decreased cytotoxicity and altered expression of 97 genes including those strongly associated with oxidative stress and cellular metabolism. By overlaying these differentially expressed genes with AHR chromatin binding, we identified 160 genes directly regulated by AHR, including hallmark AHR targets AHRR and CYP1B1 and known regulators of phenotype, development, metabolism, and function such as NCAM1, KIT, NQO1, and TXN. In summary, we define the AHR transcriptome in NK cells, propose a model of AHR and Stat3 coregulation, and identify potential pathways that may be targeted to overcome AHR-mediated immune suppression.


Subject(s)
Receptors, Aryl Hydrocarbon , Transcriptome , Cell Differentiation , Killer Cells, Natural/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction
4.
Neuro Oncol ; 22(8): 1214-1225, 2020 08 17.
Article in English | MEDLINE | ID: mdl-32152626

ABSTRACT

BACKGROUND: Recurrent pediatric medulloblastoma and ependymoma have a grim prognosis. We report a first-in-human, phase I study of intraventricular infusions of ex vivo expanded autologous natural killer (NK) cells in these tumors, with correlative studies. METHODS: Twelve patients were enrolled, 9 received protocol therapy up to 3 infusions weekly, in escalating doses from 3 × 106 to 3 × 108 NK cells/m2/infusion, for up to 3 cycles. Cerebrospinal fluid (CSF) was obtained for cellular profile, persistence, and phenotypic analysis of NK cells. Radiomic characterization on pretreatment MRI scans was performed in 7 patients, to develop a non-invasive imaging-based signature. RESULTS: Primary objectives of NK cell harvest, expansion, release, and safety of 112 intraventricular infusions of NK cells were achieved in all 9 patients. There were no dose-limiting toxicities. All patients showed progressive disease (PD), except 1 patient showed stable disease for one month at end of study follow-up. Another patient had transient radiographic response of the intraventricular tumor after 5 infusions of NK cell before progressing to PD. At higher dose levels, NK cells increased in the CSF during treatment with repetitive infusions (mean 11.6-fold). Frequent infusions of NK cells resulted in CSF pleocytosis. Radiomic signatures were profiled in 7 patients, evaluating ability to predict upfront radiographic changes, although they did not attain statistical significance. CONCLUSIONS: This study demonstrated feasibility of production and safety of intraventricular infusions of autologous NK cells. These findings support further investigation of locoregional NK cell infusions in children with brain malignancies.


Subject(s)
Brain Neoplasms , Cerebellar Neoplasms , Ependymoma , Killer Cells, Natural/transplantation , Medulloblastoma , Adolescent , Brain Neoplasms/cerebrospinal fluid , Brain Neoplasms/therapy , Cerebellar Neoplasms/cerebrospinal fluid , Cerebellar Neoplasms/therapy , Child , Ependymoma/cerebrospinal fluid , Ependymoma/drug therapy , Female , Humans , Infusions, Intraventricular , Killer Cells, Natural/immunology , Male , Medulloblastoma/cerebrospinal fluid , Medulloblastoma/therapy , Neoplasm Recurrence, Local
5.
Biochim Biophys Acta Rev Cancer ; 1873(1): 188335, 2020 01.
Article in English | MEDLINE | ID: mdl-31816350

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcriptional factor (TF) that is a member of the Per-Arnt-Sim family of proteins. AhR regulates diverse processes, including malignant transformation, hematopoietic cell development, and fate determination of immune cell lineages. Moreover, AhR forms a crucial link between innate and adaptive arms of the immune system. Malignant cells frequently evolve multiple mechanisms for suppressing tumor-specific responses, including the induction of suppressive pathways involving AhR and its metabolic byproducts in the tumor microenvironment that promote immune evasion and tumor progression. Thus, interest is high in further defining the role of AhR in carcinogenesis and immune development and regulation, particularly regarding the therapeutic interventions that unleash immune responses to cancer cells. Here, we provide an overview of the role of AhR in the regulation of innate and adaptive immune response and discuss the implications of targeting this pathway to augment the immune response in cancer patients.


Subject(s)
Gene Expression Regulation/genetics , Immune System/metabolism , Neoplasms/genetics , Receptors, Aryl Hydrocarbon/genetics , Tumor Microenvironment/genetics , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Gene Expression Regulation/immunology , Humans , Immune System/cytology , Immune System/immunology , Immune Tolerance/genetics , Immune Tolerance/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Neoplasms/immunology , Neoplasms/pathology , Receptors, Aryl Hydrocarbon/immunology , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Microenvironment/immunology
7.
J Allergy Clin Immunol ; 143(1): 346-358.e6, 2019 01.
Article in English | MEDLINE | ID: mdl-30096390

ABSTRACT

BACKGROUND: The mechanism by which natural killer (NK) cell education results in licensed NK cells with heightened effector function against missing self-targets is not known. OBJECTIVE: We sought to identify potential mechanisms of enhanced function in licensed human NK cells. METHODS: We used expanded human NK cells from killer immunoglobulin-like receptor (KIR)/HLA-genotyped donors sorted for single-KIR+ cells to generate pure populations of licensed and unlicensed NK cells. We performed proteomic and gene expression analysis of these cells before and after receptor cross-linking and performed functional and metabolic analysis before and after interference with selected metabolic pathways. We verified key findings using freshly isolated and sorted NK cells from peripheral blood. RESULTS: We confirmed that licensed human NK cells are greater in number in peripheral blood and proliferate more in vitro than unlicensed NK cells. Using high-throughput protein analysis, we found that unstimulated licensed NK cells have increased expression of the glycolytic enzyme pyruvate kinase muscle isozyme M2 and after KIR cross-linking have increased phosphorylation of the metabolic modulators p38-α and 5' adenosine monophosphate-activated protein kinase α. After cytokine expansion and activation, unlicensed NK cells depended solely on mitochondrial respiration for cytolytic function, whereas licensed NK cells demonstrated metabolic reprogramming toward glycolysis and mitochondrial-dependent glutaminolysis, leading to accumulation of glycolytic metabolites and depletion of glutamate. As such, blocking both glycolysis and mitochondrial-dependent respiration was required to suppress the cytotoxicity of licensed NK cells. CONCLUSIONS: Collectively, our data support an arming model of education in which enhanced glycolysis in licensed NK cells supports proliferative and cytotoxic capacity.


Subject(s)
Glycolysis/immunology , Lymphocyte Activation/immunology , Models, Immunological , Gene Expression Regulation/immunology , Humans , Proteomics , Receptors, KIR/immunology
8.
J Vis Exp ; (136)2018 06 14.
Article in English | MEDLINE | ID: mdl-29985369

ABSTRACT

CRISPR/Cas9 technology is accelerating genome engineering in many cell types, but so far, gene delivery and stable gene modification have been challenging in primary NK cells. For example, transgene delivery using lentiviral or retroviral transduction resulted in a limited yield of genetically-engineered NK cells due to substantial procedure-associated NK cell apoptosis. We describe here a DNA-free method for genome editing of human primary and expanded NK cells using Cas9 ribonucleoprotein complexes (Cas9/RNPs). This method allowed efficient knockout of the TGFBR2 and HPRT1 genes in NK cells. RT-PCR data showed a significant decrease in gene expression level, and a cytotoxicity assay of a representative cell product suggested that the RNP-modified NK cells became less sensitive to TGFß. Genetically modified cells could be expanded post-electroporation by stimulation with irradiated mbIL21-expressing feeder cells.


Subject(s)
CRISPR-Cas Systems/genetics , Genetic Engineering/methods , Genetic Therapy/methods , Immunotherapy/methods , Killer Cells, Natural/metabolism , Ribonucleoproteins/metabolism , Humans
9.
Clin Cancer Res ; 24(8): 1891-1904, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29363526

ABSTRACT

Purpose: mAbs are used to treat solid and hematologic malignancies and work in part through Fc receptors (FcRs) on natural killer cells (NK). However, FcR-mediated functions of NK cells from patients with cancer are significantly impaired. Identifying the mechanisms of this dysfunction and impaired response to mAb therapy could lead to combination therapies and enhance mAb therapy.Experimental Design: Cocultures of autologous NK cells and MDSC from patients with cancer were used to study the effect of myeloid-derived suppressor cells (MDSCs) on NK-cell FcR-mediated functions including antibody-dependent cellular cytotoxicity, cytokine production, and signal transduction in vitro Mouse breast cancer models were utilized to study the effect of MDSCs on antibody therapy in vivo and test the efficacy of combination therapies including a mAb and an MDSC-targeting agent.Results: MDSCs from patients with cancer were found to significantly inhibit NK-cell FcR-mediated functions including antibody-dependent cellular cytotoxicity, cytokine production, and signal transduction in a contact-independent manner. In addition, adoptive transfer of MDSCs abolished the efficacy of mAb therapy in a mouse model of pancreatic cancer. Inhibition of iNOS restored NK-cell functions and signal transduction. Finally, nonspecific elimination of MDSCs or inhibition of iNOS in vivo significantly improved the efficacy of mAb therapy in a mouse model of breast cancer.Conclusions: MDSCs antagonize NK-cell FcR-mediated function and signal transduction leading to impaired response to mAb therapy in part through nitric oxide production. Thus, elimination of MDSCs or inhibition of nitric oxide production offers a strategy to improve mAb therapy. Clin Cancer Res; 24(8); 1891-904. ©2018 AACR.


Subject(s)
Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Nitric Oxide/biosynthesis , Receptors, Fc/metabolism , Animals , Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cytokines/metabolism , Cytotoxicity, Immunologic , Disease Models, Animal , Female , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction , Xenograft Model Antitumor Assays
10.
Cancer Immunol Immunother ; 66(11): 1437-1447, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28688082

ABSTRACT

This study sought to evaluate whether myeloid-derived suppressor cells (MDSC) could be affected by chemotherapy and correlate with pathologic complete response (pCR) in breast cancer patients receiving neo-adjuvant chemotherapy. Peripheral blood levels of granulocytic (G-MDSC) and monocytic (M-MDSC) MDSC were measured by flow cytometry prior to cycle 1 and 2 of doxorubicin and cyclophosphamide and 1st and last administration of paclitaxel or paclitaxel/anti-HER2 therapy. Of 24 patients, 11, 6 and 7 patients were triple negative, HER2+ and hormone receptor+, respectively. 45.8% had pCR. Mean M-MDSC% were <1. Mean G-MDSC% and 95% confidence intervals were 0.88 (0.23-1.54), 5.07 (2.45-7.69), 9.32 (4.02-14.61) and 1.97 (0.53-3.41) at draws 1-4. The increase in G-MDSC by draw 3 was significant (p < 0.0001) in all breast cancer types. G-MDSC levels at the last draw were numerically lower in patients with pCR (1.15; 95% CI 0.14-2.16) versus patients with no pCR (2.71; 95% CI 0-5.47). There was no significant rise in G-MDSC from draw 1 to 3 in African American patients, and at draw 3 G-MDSC levels were significantly lower in African Americans versus Caucasians (p < 0.05). It was concluded that G-MDSC% increased during doxorubicin and cyclophosphamide therapy, but did not significantly differ between patients based on pathologic complete response.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/blood , Breast Neoplasms/drug therapy , Myeloid-Derived Suppressor Cells/drug effects , Adult , Black or African American , Aged , Breast Neoplasms/ethnology , Cell Count , Chemotherapy, Adjuvant , Cyclophosphamide/administration & dosage , Cytokines/blood , Doxorubicin/administration & dosage , Female , Granulocytes/drug effects , Granulocytes/pathology , Humans , Middle Aged , Monocytes/drug effects , Monocytes/pathology , Myeloid-Derived Suppressor Cells/pathology , Neoadjuvant Therapy , Paclitaxel/administration & dosage , Pilot Projects , Treatment Outcome , White People
11.
Cancer Immunol Res ; 5(9): 778-789, 2017 09.
Article in English | MEDLINE | ID: mdl-28724544

ABSTRACT

Natural killer (NK) cells are large granular lymphocytes that promote the antitumor response via communication with other cell types in the tumor microenvironment. Previously, we have shown that NK cells secrete a profile of immune stimulatory factors (e.g., IFNγ, MIP-1α, and TNFα) in response to dual stimulation with the combination of antibody (Ab)-coated tumor cells and cytokines, such as IL12. We now demonstrate that this response is enhanced in the presence of autologous monocytes. Monocyte enhancement of NK cell activity was dependent on cell-to-cell contact as determined by a Transwell assay. It was hypothesized that NK cell effector functions against Ab-coated tumor cells were enhanced via binding of MICA on monocytes to NK cell NKG2D receptors. Strategies to block MICA-NKG2D interactions resulted in reductions in IFNγ production. Depletion of monocytes in vivo resulted in decreased IFNγ production by murine NK cells upon exposure to Ab-coated tumor cells. In mice receiving trastuzumab and IL12 therapy, monocyte depletion resulted in significantly greater tumor growth in comparison to mock-depleted controls (P < 0.05). These data suggest that NK cell-monocyte interactions enhance NK cell antitumor activity in the setting of monoclonal Ab therapy for cancer. Cancer Immunol Res; 5(9); 778-89. ©2017 AACR.


Subject(s)
Breast Neoplasms/therapy , Histocompatibility Antigens Class I/immunology , Interferon-gamma/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Histocompatibility Antigens Class I/drug effects , Humans , Interleukin-12/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Mice , Monocytes/immunology , Monocytes/pathology , NK Cell Lectin-Like Receptor Subfamily K/antagonists & inhibitors , Receptors, Fc/administration & dosage , Receptors, Fc/immunology , Trastuzumab/administration & dosage , Trastuzumab/immunology
12.
Oncoimmunology ; 5(9): e1214787, 2016.
Article in English | MEDLINE | ID: mdl-27757311

ABSTRACT

Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of early myeloid cells that accumulate in the blood and tumors of patients with cancer. MDSC play a critical role during tumor evasion and promote immune suppression through variety of mechanisms, such as the generation of reactive oxygen and nitrogen species (ROS and RNS) and cytokines. AMPactivated protein kinase (AMPK) is an evolutionarily conserved serine/threonine kinase that regulates energy homeostasis and metabolic stress. However, the role of AMPK in the regulation of MDSC function remains largely unexplored. This study was designed to investigate whether treatment of MDSC with OSU-53, a PPAR-inactive derivative that stimulates AMPK kinase, can modulate MDSC function. Our results demonstrate that OSU-53 treatment increases the phosphorylation of AMPK, significantly reduces nitric oxide production, inhibits MDSC migration, and reduces the levels of IL-6 in murine MDSC cell line (MSC2 cells). OSU53 treatment mitigated the immune suppressive functions of murine MDSC, promoting T-cell proliferation. Although OSU-53 had a modest effect on tumor growth in mice inoculated with EMT-6 cells, importantly, administration of OSU53 significantly (p < 0.05) reduced the levels of MDSC in the spleens and tumors. Furthermore, mouse MDSC from EMT-6 tumor-bearing mice and human MDSC isolated from melanoma patients treated with OSU-53 showed a significant reduction in the expression of immune suppressive genes iNOS and arginase. In summary, these results demonstrate a novel role of AMPK in the regulation of MDSC functions and provide a rationale of combining OSU-53 with immune checkpoint inhibitors to augment their response in cancer patients.

13.
Mol Cancer Ther ; 15(9): 2244-50, 2016 09.
Article in English | MEDLINE | ID: mdl-27458141

ABSTRACT

mAbs can induce antibody-dependent cellular cytotoxicity (ADCC) via the innate immune system's ability to recognize mAb-coated cancer cells and activate immune effector cells. Lenalidomide is an immunomodulatory agent with the capacity to stimulate immune cell cytokine production and ADCC activity. This phase I trial evaluated the combination of cetuximab with lenalidomide for the treatment of advanced colorectal and head and neck squamous cell cancers (HNSCC). This trial included patients with advanced colorectal cancer or HNSCC. Treatment consisted of cetuximab 500 mg/m(2) i.v. every two weeks with lenalidomide given orally days 1-21 on a 28-day cycle. Three dose levels of lenalidomide were evaluated (15, 20, 25 mg). Correlative studies included measurement of ADCC, FcγRIIIA polymorphism genotyping, measurement of serum cytokine levels, and flow cytometric analysis of immune cell subtypes. Twenty-two patients were enrolled (19 colorectal cancer, 3 HNSCC). Fatigue was the only dose-limiting toxicity. One partial response was observed and 8 patients had stable disease at least 12 weeks. The recommended phase II dose is cetuximab 500 mg/m(2) with lenalidomide 25 mg daily, days 1-21. Correlative studies demonstrated a dose-dependent increase in natural killer cytotoxic activity with increasing doses of lenalidomide. Cetuximab and lenalidomide were well tolerated. There was a lenalidomide dose-dependent increase in ADCC with higher activity in patients enrolled in cohort 3 than those enrolled in cohorts 1/2. Although response was not a primary endpoint, there was evidence of antitumor activity for the combination therapy. Further investigation of lenalidomide as an immunomodulator in solid tumors is warranted. Mol Cancer Ther; 15(9); 2244-50. ©2016 AACR.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cetuximab/immunology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/immunology , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cetuximab/administration & dosage , Colorectal Neoplasms/pathology , Cytokines/metabolism , Female , Head and Neck Neoplasms/pathology , Humans , Lenalidomide , Male , Middle Aged , Neoplasm Staging , Retreatment , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Treatment Outcome
14.
Cancer Res ; 76(8): 2125-36, 2016 04 15.
Article in English | MEDLINE | ID: mdl-26880800

ABSTRACT

Myeloid-derived suppressor cells (MDSC) are a heterogeneous group of immature myeloid cells that expand in tumor-bearing hosts in response to soluble factors produced by tumor and stromal cells. MDSC expansion has been linked to loss of immune effector cell function and reduced efficacy of immune-based cancer therapies, highlighting the MDSC population as an attractive therapeutic target. Ibrutinib, an irreversible inhibitor of Bruton's tyrosine kinase (BTK) and IL2-inducible T-cell kinase (ITK), is in clinical use for the treatment of B-cell malignancies. Here, we report that BTK is expressed by murine and human MDSCs, and that ibrutinib is able to inhibit BTK phosphorylation in these cells. Treatment of MDSCs with ibrutinib significantly impaired nitric oxide production and cell migration. In addition, ibrutinib inhibited in vitro generation of human MDSCs and reduced mRNA expression of indolamine 2,3-dioxygenase, an immunosuppressive factor. Treatment of mice bearing EMT6 mammary tumors with ibrutinib resulted in reduced frequency of MDSCs in both the spleen and tumor. Ibrutinib treatment also resulted in a significant reduction of MDSCs in wild-type mice bearing B16F10 melanoma tumors, but not in X-linked immunodeficiency mice (XID) harboring a BTK mutation, suggesting that BTK inhibition plays an important role in the observed reduction of MDSCs in vivo Finally, ibrutinib significantly enhanced the efficacy of anti-PD-L1 (CD274) therapy in a murine breast cancer model. Together, these results demonstrate that ibrutinib modulates MDSC function and generation, revealing a potential strategy for enhancing immune-based therapies in solid malignancies. Cancer Res; 76(8); 2125-36. ©2016 AACR.


Subject(s)
Myeloid-Derived Suppressor Cells/enzymology , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/metabolism , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Xenograft Model Antitumor Assays , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase , Animals , Apoptosis , Cell Line, Tumor , Cytokines/biosynthesis , Gene Expression , Humans , Immunotherapy , Mice , Piperidines
15.
BMC Med Genomics ; 8: 66, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26470881

ABSTRACT

BACKGROUND: Traditionally, the CD56(dim)CD16(+) subset of Natural Killer (NK) cells has been thought to mediate cellular cytotoxicity with modest cytokine secretion capacity. However, studies have suggested that this subset may exert a more diverse array of immunological functions. There exists a lack of well-developed functional models to describe the behavior of activated NK cells, and the interactions between signaling pathways that facilitate effector functions are not well understood. In the present study, a combination of genome-wide microarray analyses and systems-level bioinformatics approaches were utilized to elucidate the transcriptional landscape of NK cells activated via interactions with antibody-coated targets in the presence of interleukin-12 (IL-12). METHODS: We conducted differential gene expression analysis of CD56(dim)CD16(+) NK cells following FcR stimulation in the presence or absence of IL-12. Next, we functionally characterized gene sets according to patterns of gene expression and validated representative genes using RT-PCR. IPA was utilized for biological pathway analysis, and an enriched network of interacting genes was generated using GeneMANIA. Furthermore, PAJEK and the HITS algorithm were employed to identify important genes in the network according to betweeness centrality, hub, and authority node metrics. RESULTS: Analyses revealed that CD56(dim)CD16(+) NK cells co-stimulated via the Fc receptor (FcR) and IL-12R led to the expression of a unique set of genes, including genes encoding cytotoxicity receptors, apoptotic proteins, intracellular signaling molecules, and cytokines that may mediate enhanced cytotoxicity and interactions with other immune cells within inflammatory tissues. Network analyses identified a novel set of connected key players, BATF, IRF4, TBX21, and IFNG, within an integrated network composed of differentially expressed genes in NK cells stimulated by various conditions (immobilized IgG, IL-12, or the combination of IgG and IL-12). CONCLUSIONS: These results are the first to address the global mechanisms by which NK cells mediate their biological functions when encountering antibody-coated targets within inflammatory sites. Moreover, this study has identified a set of high-priority targets for subsequent investigation into strategies to combat cancer by enhancing the anti-tumor activity of CD56(dim)CD16(+) NK cells.


Subject(s)
Interleukin-12/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Receptors, Fc/metabolism , Transcriptome/drug effects , Gene Regulatory Networks/drug effects , Genomics , Humans , Immunoglobulin G/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Oligonucleotide Array Sequence Analysis
16.
J Immunol ; 195(5): 1995-2005, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26238487

ABSTRACT

Sorafenib is an oral multikinase inhibitor that was originally developed as a Raf kinase inhibitor. We hypothesized that sorafenib would also have inhibitory effects on cytokine signaling pathways in immune cells. PBMCs from normal donors were treated with varying concentrations of sorafenib and stimulated with IFN-α or IL-2. Phosphorylation of STAT1 and STAT5 was measured by flow cytometry and confirmed by immunoblot analysis. Changes in IFN-α- and IL-2-stimulated gene expression were measured by quantitative PCR, and changes in cytokine production were evaluated by ELISA. Cryopreserved PBMCs were obtained from cancer patients before and after receiving 400 mg sorafenib twice daily. Patient PBMCs were thawed, stimulated with IL-2 or IFN-α, and evaluated for phosphorylation of STAT1 and STAT5. Pretreatment of PBMCs with 10 µM sorafenib decreased STAT1 and STAT5 phosphorylation after treatment with IFN-α or IL-2. This inhibitory effect was observed in PBMCs from healthy donors over a range of concentrations of sorafenib (5-20 µM), IL-2 (2-24 nM), and IFN-α (10(1)-10(6) U/ml). This effect was observed in immune cell subsets, including T cells, B cells, NK cells, regulatory T cells, and myeloid-derived suppressor cells. Pretreatment with sorafenib also inhibited PBMC expression of IFN-α- and IL-2-regulated genes and inhibited NK cell production of IFN-γ, RANTES, MIP1-α, and MIG in response to IFN-α stimulation. PBMCs from patients receiving sorafenib therapy showed decreased responsiveness to IL-2 and IFN-α treatment. Sorafenib is a Raf kinase inhibitor that could have off-target effects on cytokine-induced signal transduction in immune effector cells.


Subject(s)
Janus Kinase 1/metabolism , Leukocytes, Mononuclear/drug effects , Protein Kinase Inhibitors/pharmacology , STAT1 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Cells, Cultured , Dose-Response Relationship, Drug , Flow Cytometry , Gene Expression/drug effects , Humans , Immunoblotting , Interferon-alpha/pharmacology , Interleukin-2/pharmacology , K562 Cells , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocytes/drug effects , Lymphocytes/metabolism , Mice, Inbred BALB C , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenylurea Compounds/pharmacology , Phosphorylation/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Sorafenib , Thyroid Neoplasms/blood , Thyroid Neoplasms/drug therapy , raf Kinases/antagonists & inhibitors , raf Kinases/metabolism
17.
Biochim Biophys Acta ; 1846(1): 55-65, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24727385

ABSTRACT

The immune system has evolved mechanisms to protect the host from the deleterious effects of inflammation. The generation of immune suppressive cells like myeloid derived suppressor cells (MDSCs) that can counteract T cell responses represents one such strategy. There is an accumulation of immature myeloid cells or MDSCs in bone marrow (BM) and lymphoid organs under pathological conditions such as cancer. MDSCs represent a population of heterogeneous myeloid cells comprising of macrophages, granulocytes and dendritic cells that are at early stages of development. Although, the precise signaling pathways and molecular mechanisms that lead to MDSC generation and expansion in cancer remains to be elucidated. It is widely believed that perturbation of signaling pathways involved during normal hematopoietic and myeloid development under pathological conditions such as tumorogenesis contributes to the development of suppressive myeloid cells. In this review we discuss the role played by key signaling pathways such as PI3K, Ras, Jak/Stat and TGFb during myeloid development and how their deregulation under pathological conditions can lead to the generation of suppressive myeloid cells or MDSCs. Targeting these pathways should help in elucidating mechanisms that lead to the expansion of MDSCs in cancer and point to methods for eliminating these cells from the tumor microenvironment.


Subject(s)
Myeloid Cells/physiology , Neoplasms/immunology , Animals , Cell Transformation, Neoplastic/immunology , Cytokines/physiology , Dendritic Cells/physiology , Granulocytes/physiology , Humans , Macrophages/physiology , Neoplasms/prevention & control , Signal Transduction/physiology
18.
Nat Cell Biol ; 14(11): 1192-202, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23064266

ABSTRACT

The endocycle is a variant cell cycle consisting of successive DNA synthesis and gap phases that yield highly polyploid cells. Although essential for metazoan development, relatively little is known about its control or physiologic role in mammals. Using lineage-specific cre mice we identified two opposing arms of the E2F program, one driven by canonical transcription activation (E2F1, E2F2 and E2F3) and the other by atypical repression (E2F7 and E2F8), that converge on the regulation of endocycles in vivo. Ablation of canonical activators in the two endocycling tissues of mammals, trophoblast giant cells in the placenta and hepatocytes in the liver, augmented genome ploidy, whereas ablation of atypical repressors diminished ploidy. These two antagonistic arms coordinate the expression of a unique G2/M transcriptional program that is critical for mitosis, karyokinesis and cytokinesis. These results provide in vivo evidence for a direct role of E2F family members in regulating non-traditional cell cycles in mammals.


Subject(s)
Cell Cycle/physiology , E2F Transcription Factors/metabolism , Animals , Cell Cycle/genetics , Chromatin Immunoprecipitation , E2F Transcription Factors/genetics , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/metabolism , E2F3 Transcription Factor/genetics , E2F3 Transcription Factor/metabolism , E2F7 Transcription Factor/genetics , E2F7 Transcription Factor/metabolism , Female , Flow Cytometry , Giant Cells/cytology , Giant Cells/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Immunohistochemistry , Mice , Microscopy, Confocal , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Pregnancy , Repressor Proteins/genetics , Repressor Proteins/metabolism , Trophoblasts/metabolism
19.
Cancer Res ; 72(3): 604-15, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22158945

ABSTRACT

S100A7/psoriasin, a member of the epidermal differentiation complex, is widely overexpressed in invasive estrogen receptor (ER)α-negative breast cancers. However, it has not been established whether S100A7 contributes to breast cancer growth or metastasis. Here, we report the consequences of its expression on inflammatory pathways that impact breast cancer growth. Overexpression of human S100A7 or its murine homologue mS100a7a15 enhanced cell proliferation and upregulated various proinflammatory molecules in ERα-negative breast cancer cells. To examine in vivo effects, we generated mice with an inducible form of mS100a7a15 (MMTV-mS100a7a15 mice). Orthotopic implantation of MVT-1 breast tumor cells into the mammary glands of these mice enhanced tumor growth and metastasis. Compared with uninduced transgenic control mice, the mammary glands of mice where mS100a7a15 was induced exhibited increased ductal hyperplasia and expression of molecules involved in proliferation, signaling, tissue remodeling, and macrophage recruitment. Furthermore, tumors and lung tissues obtained from these mice showed further increases in prometastatic gene expression and recruitment of tumor-associated macrophages (TAM). Notably, in vivo depletion of TAM inhibited the effects of mS100a7a15 induction on tumor growth and angiogenesis. Furthermore, introduction of soluble hS100A7 or mS100a7a15 enhanced chemotaxis of macrophages via activation of RAGE receptors. In summary, our work used a powerful new model system to show that S100A7 enhances breast tumor growth and metastasis by activating proinflammatory and metastatic pathways.


Subject(s)
Inflammation/genetics , Mammary Neoplasms, Experimental/genetics , S100 Proteins/genetics , Signal Transduction/genetics , Animals , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Chemokines/genetics , Chemokines/metabolism , Chemotaxis/genetics , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Inflammation/metabolism , Inflammation/pathology , Macrophages/metabolism , Macrophages/pathology , Male , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Reverse Transcriptase Polymerase Chain Reaction , S100 Calcium Binding Protein A7 , S100 Proteins/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
20.
J Biol Chem ; 286(6): 4783-95, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21115501

ABSTRACT

Hematopoietic development involves the coordinated activity of differentiation and cell cycle regulators. In current models of mammalian cell cycle control, E2f activators (E2f1, E2f2, and E2f3) are portrayed as the ultimate transcriptional effectors that commit cells to enter and progress through S phase. Using conditional gene knock-out strategies, we show that E2f1-3 are not required for the proliferation of early myeloid progenitors. Rather, these E2fs are critical for cell survival and proliferation at two distinct steps of myeloid development. First, E2f1-3 are required as transcriptional repressors for the survival of CD11b(+) myeloid progenitors, and then they are required as activators for the proliferation of CD11b(+) macrophages. In bone marrow macrophages, we show that E2f1-3 respond to CSF1-Myc mitogenic signals and serve to activate E2f target genes and promote their proliferation. Together, these findings expose dual functions for E2f1-3 at distinct stages of myeloid development in vivo, first as repressors in cell survival and then as activators in cell proliferation. In summary, this work places E2f1-3 in a specific signaling cascade that is critical for myeloid development in vivo.


Subject(s)
Cell Differentiation/physiology , E2F1 Transcription Factor/metabolism , E2F2 Transcription Factor/metabolism , E2F3 Transcription Factor/metabolism , Myeloid Progenitor Cells/metabolism , S Phase/physiology , Animals , CD11b Antigen/genetics , CD11b Antigen/metabolism , Cell Survival/physiology , E2F1 Transcription Factor/genetics , E2F2 Transcription Factor/genetics , E2F3 Transcription Factor/genetics , Gene Knockout Techniques , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Knockout , NIH 3T3 Cells , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...