Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Immunol ; 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38921973

ABSTRACT

Stroke is one of the leading causes of death and long-term disabilities worldwide. In addition to interruption of blood flow, inflammation is widely recognized as an important factor mediating tissue destruction in stroke. Depending on their phenotype, microglia, the main leukocytes in the CNS, are capable of either causing further tissue damage or promoting brain restoration after stroke. ß2-integrins are cell adhesion molecules that are constitutively expressed on microglia. The function of ß2-integrins has been investigated extensively in animal models of ischemic stroke, but their role in hemorrhagic stroke is currently poorly understood. We show in this study that dysfunction of ß2-integrins is associated with improved functional outcome and decreased inflammatory cytokine expression in the brain in a mouse model of hemorrhagic stroke. Furthermore, ß2-integrins affect microglial phenotype and cytokine responses in vivo. Therefore, our findings suggest that targeting ß2-integrins in hemorrhagic stroke may be beneficial.

2.
Oncoimmunology ; 13(1): 2369373, 2024.
Article in English | MEDLINE | ID: mdl-38915784

ABSTRACT

Dendritic cells (DCs) are the main antigen presenting cells of the immune system and are essential for anti-tumor responses. DC-based immunotherapies are used in cancer treatment, but their functionality is not optimized and their clinical efficacy is currently limited. Approaches to improve DC functionality in anti-tumor immunity are therefore required. We have previously shown that the loss of ß2-integrin-mediated adhesion leads to epigenetic reprogramming of bone marrow-derived DCs (BM-DCs), resulting in an increased expression of costimulatory markers (CD86, CD80, and CD40), cytokines (IL-12) and the chemokine receptor CCR7. We now show that the loss of ß2-integrin-mediated adhesion of BM-DCs also leads to a generally suppressed metabolic profile, with reduced metabolic rate, decreased ROS production, and lowered glucose uptake in cells. The mRNA levels of glycolytic enzymes and glucose transporters were reduced, indicating transcriptional regulation of the metabolic phenotype. Surprisingly, although signaling through a central regulator of immune cell metabolisms, the mechanistic target of rapamycin (mTOR), was increased in BM-DCs with dysfunctional integrins, rapamycin treatment revealed that mTOR signaling was not involved in suppressing DC metabolism. Instead, bioinformatics and functional analyses showed that the Ikaros transcription factor may be involved in regulating the metabolic profile of non-adhesive DCs. Inversely, we found that induction of metabolic stress through treatment of cells with low levels of an inhibitor of glycolysis, 2-deoxyglucose (2DG), led to increased BM-DC activation. Specifically, 2DG treatment led to increased levels of Il-12 and Ccr7 mRNA, increased production of IL-12, increased levels of cell surface CCR7 and increased in vitro migration and T cell activation potential. Furthermore, 2DG treatment led to increased histone methylation in cells (H3K4me3, H3K27me3), indicating metabolic reprogramming. Finally, metabolic stress induced by 2DG treatment led to improved BM-DC-mediated anti-tumor responses in vivo in a melanoma cancer model, B16-OVA. In conclusion, our results indicate a role for ß2-integrin-mediated adhesion in regulating a novel type of metabolic reprogramming of DCs and DC-mediated anti-tumor responses, which may be targeted to enhance DC-mediated anti-tumor responses in cancer immunotherapy.


Subject(s)
CD18 Antigens , Dendritic Cells , Dendritic Cells/metabolism , Dendritic Cells/immunology , Animals , Mice , CD18 Antigens/metabolism , CD18 Antigens/genetics , Mice, Inbred C57BL , Cell Adhesion , Receptors, CCR7/metabolism , Receptors, CCR7/genetics , Melanoma, Experimental/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/genetics , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Humans , Metabolic Reprogramming
3.
Cancer Immunol Res ; 9(11): 1354-1369, 2021 11.
Article in English | MEDLINE | ID: mdl-34561280

ABSTRACT

Dendritic cells (DC), the classic antigen-presenting cells of the immune system, switch from an adhesive, phagocytic phenotype in tissues, to a mature, nonadhesive phenotype that enables migration to lymph nodes to activate T cells and initiate antitumor responses. Monocyte-derived DCs are used in cancer immunotherapy, but their clinical efficacy is limited. Here, we show that cultured bone marrow-derived DCs (BM-DC) expressing dysfunctional ß2-integrin adhesion receptors displayed enhanced tumor rejection capabilities in B16.OVA and B16-F10 melanoma models. This was associated with an increased CD8+ T-cell response. BM-DCs expressing dysfunctional ß2-integrins or manipulated to disrupt integrin adhesion or integrin/actin/nuclear linkages displayed spontaneous maturation in ex vivo cultures (increased costimulatory marker expression, IL12 production, and 3D migration capabilities). This spontaneous maturation was associated with an altered DC epigenetic/transcriptional profile, including a global increase in chromatin accessibility and H3K4me3/H3K27me3 histone methylation. Genome-wide analyses showed that H3K4me3 methylation was increased on DC maturation genes, such as CD86, Il12, Ccr7, and Fscn1, and revealed a role for a transcription factor network involving Ikaros and RelA in the integrin-regulated phenotype of DCs. Manipulation of the integrin-regulated epigenetic landscape in wild-type ex vivo-cultured BM-DCs enhanced their functionality in tumor rejection in vivo. Thus, ß2-integrin-mediated adhesion to the extracellular environment plays an important role in restricting DC maturation and antitumor responses through regulation of the cellular epigenetic and transcriptional landscape. Targeting ß2-integrins could therefore be a new strategy to improve the performance of current DC-based cancer immunotherapies.


Subject(s)
CD18 Antigens/metabolism , Epigenesis, Genetic/genetics , Neoplasms/immunology , Animals , Cell Differentiation , Dendritic Cells/immunology , Humans , Mice , Signal Transduction
4.
Immunol Cell Biol ; 99(1): 65-83, 2021 01.
Article in English | MEDLINE | ID: mdl-32748462

ABSTRACT

Type 2 innate lymphoid cells (ILC2s) are important producers of type 2 cytokines whose role in hematological cancers remains unclear. ILC2s are a heterogeneous population encompassing distinct subsets with different tissue localization and cytokine responsiveness. In this study, we investigated the role of bone marrow (BM) ILC2s and interleukin (IL)-33-stimulated ILC2s in multiple myeloma, a plasma cell malignancy that develops in the BM. We found that myeloma growth was associated with phenotypic and functional alterations of BM ILC2s, characterized by an increased expression of maturation markers and reduced cytokine response to IL-2/IL-33. We identified a population of KLRG1hi ILC2s that preferentially accumulated in the liver and spleen of Il2rg-/- Rag2-/- mice reconstituted with BM ILC2s. A similar population of KLRG1hi ILC2s was observed in the blood, liver and spleen of IL-33-treated wild-type mice. The presence of KLRG1hi ILC2s in ILC2-reconstituted Il2rg-/- Rag2-/- mice or in IL-33-treated wild-type mice was associated with increased eosinophil numbers but had no effect on myeloma progression. Interestingly, while decreased myeloma growth was observed following treatment of Rag-deficient mice with the type 1 cytokines IL-12 and IL-18, this protection was reversed when mice received a combined treatment of IL-33 together with IL-12 and IL-18. In summary, our data indicate that IL-33 treatment induces a population of circulating inflammatory KLRG1hi ILC2s and inhibits type 1 immunity against multiple myeloma. These results argue against therapeutic administration of IL-33 to myeloma patients.


Subject(s)
Immunity, Innate , Multiple Myeloma , Animals , Cytokines , Humans , Interleukin-33 , Lectins, C-Type , Lymphocytes , Mice , Multiple Myeloma/drug therapy , Receptors, Immunologic
5.
JCI Insight ; 52019 06 13.
Article in English | MEDLINE | ID: mdl-31194697

ABSTRACT

Immunotherapy holds promise for multiple myeloma (MM) patients but little is known about how MM-induced immunosuppression influences response to therapy. Here, we investigated the impact of disease progression on immunotherapy efficacy in the Vk*MYC mouse model. Treatment with agonistic anti-CD137 (4-1BB) mAbs efficiently protected mice when administered early but failed to contain MM growth when delayed more than three weeks after Vk*MYC tumor cell challenge. The quality of CD8+ T cell response to CD137 stimulation was not altered by the presence of MM, but CD8+ T cell numbers were profoundly reduced at the time of treatment. Our data suggest that an insufficient ratio of CD8+ T cells over MM cells (CD8/MM) accounts for the loss of anti-CD137 mAb efficacy. We established serum M-protein levels prior to therapy as a predictive factor of response. Moreover, we developed an in silico model to capture the dynamic interactions between CD8+ T cells and MM cells. Finally, we explored two methods to improve the CD8/MM ratio: anti-CD137 mAb immunotherapy combined with Treg-depletion or administered after chemotherapy treatment with cyclophosphamide or melphalan efficiently reduced MM burden and prolonged survival. Altogether, our data indicate that consolidation treatment with anti-CD137 mAbs might prevent MM relapse.


Subject(s)
4-1BB Ligand/metabolism , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Immunotherapy/methods , Multiple Myeloma/drug therapy , Animals , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Antineoplastic Agents/therapeutic use , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Myeloma/pathology , T-Lymphocytes, Regulatory
6.
Front Immunol ; 10: 1078, 2019.
Article in English | MEDLINE | ID: mdl-31231358

ABSTRACT

The immune system and cancer have a complex relationship with the immune system playing a dual role in tumor development. The effector cells of the immune system can recognize and kill malignant cells while immune system-mediated inflammation can also promote tumor growth and regulatory cells suppress the anti-tumor responses. In the center of all anti-tumor responses is the ability of the immune cells to migrate to the tumor site and to interact with each other and with the malignant cells. Cell adhesion molecules including receptors of the immunoglobulin superfamily and integrins are of crucial importance in mediating these processes. Particularly integrins play a vital role in regulating all aspects of immune cell function including immune cell trafficking into tissues, effector cell activation and proliferation and the formation of the immunological synapse between immune cells or between immune cell and the target cell both during homeostasis and during inflammation and cancer. In this review we discuss the molecular mechanisms regulating integrin function and the role of integrins and other cell adhesion molecules in immune responses and in the tumor microenvironment. We also describe how malignant cells can utilize cell adhesion molecules to promote tumor growth and metastases and how these molecules could be targeted in cancer immunotherapy.


Subject(s)
Cell Adhesion Molecules/immunology , Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Dendritic Cells/immunology , Humans , T-Lymphocytes/immunology
7.
Front Immunol ; 10: 1138, 2019.
Article in English | MEDLINE | ID: mdl-31191527

ABSTRACT

ß2-integrins are essential for immune system function because they mediate immune cell adhesion and signaling. Consequently, a loss of ß2-integrin expression or function causes the immunodeficiency disorders, Leukocyte Adhesion Deficiency (LAD) type I and III. LAD-III is caused by mutations in an important integrin regulator, kindlin-3, but exactly how kindlin-3 regulates leukocyte adhesion has remained incompletely understood. Here we demonstrate that mutation of the kindlin-3 binding site in the ß2-integrin (TTT/AAA-ß2-integrin knock-in mouse/KI) abolishes activation of the actin-regulated myocardin related transcription factor A/serum response factor (MRTF-A/SRF) signaling pathway in dendritic cells and MRTF-A/SRF-dependent gene expression. We show that Ras homolog gene family, member A (RhoA) activation and filamentous-actin (F-actin) polymerization is abolished in murine TTT/AAA-ß2-integrin KI dendritic cells, which leads to a failure of MRTF-A to localize to the cell nucleus to coactivate genes together with SRF. In addition, we show that dendritic cell gene expression, adhesion and integrin-mediated traction forces on ligand coated surfaces is dependent on the MRTF-A/SRF signaling pathway. The participation of ß2-integrin and kindlin-3-mediated cell adhesion in the regulation of the ubiquitous MRTF-A/SRF signaling pathway in immune cells may help explain the role of ß2-integrin and kindlin-3 in integrin-mediated gene regulation and immune system function.


Subject(s)
CD18 Antigens/metabolism , Dendritic Cells/metabolism , Gene Expression Profiling/methods , Serum Response Factor/metabolism , Trans-Activators/metabolism , Animals , Biomechanical Phenomena , CD18 Antigens/genetics , Cell Adhesion/genetics , Cell Movement/genetics , Cells, Cultured , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Dendritic Cells/cytology , Gene Ontology , Gene Regulatory Networks , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Serum Response Factor/genetics , Signal Transduction/genetics , Trans-Activators/genetics , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
8.
Blood ; 132(16): 1689-1694, 2018 10 18.
Article in English | MEDLINE | ID: mdl-29986909

ABSTRACT

Immune-based therapies hold promise for the treatment of multiple myeloma (MM), but so far, immune checkpoint blockade targeting programmed cell death protein 1 has not proven effective as single agent in this disease. T-cell immunoglobulin and ITIM domains (TIGIT) is another immune checkpoint receptor known to negatively regulate T-cell functions. In this study, we investigated the therapeutic potential of TIGIT blockade to unleash immune responses against MM. We observed that, in both mice and humans, MM progression was associated with high levels of TIGIT expression on CD8+ T cells. TIGIT+ CD8+ T cells from MM patients exhibited a dysfunctional phenotype characterized by decreased proliferation and inability to produce cytokines in response to anti-CD3/CD28/CD2 or myeloma antigen stimulation. Moreover, when challenged with Vk*MYC mouse MM cells, TIGIT-deficient mice showed decreased serum monoclonal immunoglobulin protein levels associated with reduced tumor burden and prolonged survival, indicating that TIGIT limits antimyeloma immune responses. Importantly, blocking TIGIT using monoclonal antibodies increased the effector function of MM patient CD8+ T cells and suppressed MM development. Altogether our data provide evidence for an immune-inhibitory role of TIGIT in MM and support the development of TIGIT-blocking strategies for the treatment of MM patients.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD8-Positive T-Lymphocytes/immunology , Multiple Myeloma/prevention & control , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, Immunologic/antagonists & inhibitors , Animals , CD8-Positive T-Lymphocytes/drug effects , Cells, Cultured , Humans , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Myeloma/etiology , Multiple Myeloma/pathology , Programmed Cell Death 1 Receptor/immunology , Receptors, Immunologic/metabolism , Receptors, Immunologic/physiology
9.
Cancer Immunol Res ; 6(8): 978-987, 2018 08.
Article in English | MEDLINE | ID: mdl-29921599

ABSTRACT

Tumor-induced immunosuppression is mediated through various mechanisms including engagement of immune checkpoint receptors on effector cells, function of immunoregulatory cells such as regulatory T cells and myeloid-derived suppressor cells, and deployment of immunosuppressive cytokines such as TGFß and IL10. IL23 is a cytokine that negatively affects antitumor immunity. In this study, we investigated whether IL23-deficient (IL23p19-/-) and IL23R-deficient (IL23R-/-) mice phenocopied each other, with respect to their tumor control. We found that IL23R-/- mice had significantly fewer lung metastases compared with IL23p19-/- mice across three different experimental lung metastasis models (B16F10, LWT1, and RM-1). Similarly, IL23R blocking antibodies were more effective than antibodies neutralizing IL23 in suppressing experimental lung metastases. The antimetastatic activity of anti-IL23R was dependent on NK cells and IFNγ but independent of CD8+ T cells, CD4+ T cells, activating Fc receptors, and IL12. Furthermore, our data suggest this increased antitumor efficacy was due to an increase in the proportion of IFNγ-producing NK cells in the lungs of B16F10 tumor-bearing mice. Anti-IL23R, but not anti-IL23p19, partially suppressed lung metastases in tumor-bearing mice neutralized for IL12p40. Collectively, our data imply that IL23R has tumor-promoting effects that are partially independent of IL23p19. Blocking IL23R may be more effective than neutralizing IL23 in the suppression of tumor metastases. Cancer Immunol Res; 6(8); 978-87. ©2018 AACR.


Subject(s)
Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Receptors, Interleukin/antagonists & inhibitors , Animals , Immunotherapy/methods , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-23/antagonists & inhibitors , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin/deficiency , Receptors, Interleukin/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Oncoimmunology ; 7(7): e1445949, 2018.
Article in English | MEDLINE | ID: mdl-29900061

ABSTRACT

Multiple non-redundant immunosuppressive pathways co-exist in the tumor microenvironment and their co-targeting can increase clinical responses. Indeed, concurrent blockade of CTLA-4 and PD-1 in patients with advanced melanoma increased clinical responses over monotherapy alone although the frequency and severity of immune related adverse events (irAEs) also increased. Nevertheless, a substantial number of patients still display an innate resistance phenotype and are unresponsive to current approved immunotherapies even when utilized in combination. In this study, we generated Pdcd1-/-CD96-/- and Tigit-/-CD96-/- mice to investigate how loss of CD96 in combination with PD-1 or TIGIT impacts on immune homeostasis and hence the potential of inducing immune related toxicities following co-targeting of these pairs of receptors. The ability of Pdcd1-/-CD96-/- and Tigit-/-CD96-/- mice to suppress primary tumor growth was also assessed using the MC38 colon carcinoma and SM1WT1 BRAF-mutated melanoma tumor models. Both Pdcd1-/-CD96-/- or Tigit-/-CD96-/- mice displayed no overt perturbations in immune homeostasis over what was previously reported with Pdcd1-/- or Tigit-/- mice even when aged for 22 months. Interestingly, increased suppression of subcutaneous tumor growth and complete responses was seen in Pdcd1-/-CD96-/- mice compared to Pdcd1-/- or CD96-/- mice depending upon the tumor model. In contrast, in these models, growth suppression in Tigit-/-CD96-/- were similar to Tigit-/- or CD96-/- . This enhanced anti-tumor efficacy of Pdcd1-/-CD96-/- appeared to be due to favorable changes in the ratio of CD8+ T cells to T regulatory cells or CD11b+GR-1hi myeloid cells in the tumor microenvironment. Co-targeting CD96 and PD-1 may increase anti-tumor immunity over targeting PD-1 alone and potentially not induce serious immune-related toxicities and thus appears a promising strategy for clinical development.

11.
Oncoimmunology ; 7(6): e1431088, 2018.
Article in English | MEDLINE | ID: mdl-29872559

ABSTRACT

Receptor activator of NF-κB ligand (RANKL) and its receptor RANK, are members of the tumor necrosis factor and receptor superfamilies, respectively. Antibodies targeting RANKL have recently been evaluated in combination with anti-CTLA4 in case reports of human melanoma and mouse models of cancer. However, the efficacy of anti-RANKL in combination with antibodies targeting other immune checkpoint receptors such as PD1 has not been reported. In this study, we demonstrated that blockade of RANKL improves anti-metastatic activity of antibodies targeting PD1/PD-L1 and improves subcutaneous growth suppression in mouse models of melanoma, prostate and colon cancer. Suppression of experimental lung metastasis following combination anti-RANKL with anti-PD1 requires NK cells and IFN-γ, whereas subcutaneous tumor growth suppression with this combination therapy is attenuated in the absence of T cells and IFN-γ. Furthermore, addition of anti-RANKL to anti-PD1 and anti-CTLA4 resulted in superior anti-tumor responses, irrespective of the ability of anti-CTLA4 isotype to engage activating FcR, and concurrent or delayed RANKL blockade was most effective. Early-during-treatment assessment reveals this triple combination therapy compared to dual anti-PD1 and anti-CTLA4 combination therapy further increased the proportion of tumor-infiltrating CD4+ and CD8+ T cells that can produce both IFN-γ and TNF. Finally, RANKL expression appears to identify tumor-specific CD8+ T cells expressing higher levels of PD1 which can be modulated by anti-PD1. These data set the scene for clinical evaluation of denosumab use in patients receiving contemporary immune checkpoint blockade.

12.
Stand Genomic Sci ; 12: 87, 2017.
Article in English | MEDLINE | ID: mdl-29276572

ABSTRACT

Bacteria of the genus Pectobacterium are economically important plant pathogens that cause soft rot disease on a wide variety of plant species. Here, we report the genome sequence of Pectobacterium carotovorum strain SCC1, a Finnish soft rot model strain isolated from a diseased potato tuber in the early 1980's. The genome of strain SCC1 consists of one circular chromosome of 4,974,798 bp and one circular plasmid of 5524 bp. In total 4451 genes were predicted, of which 4349 are protein coding and 102 are RNA genes.

13.
Clin Cancer Res ; 23(19): 5789-5801, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28634284

ABSTRACT

Purpose: Novel partners for established immune checkpoint inhibitors in the treatment of cancer are needed to address the problems of primary and acquired resistance. The efficacy of combination RANKL and CTLA4 blockade in antitumor immunity has been suggested by recent case reports in melanoma. Here, we provide a rationale for this combination in mouse models of cancer.Experimental Design: The efficacy and mechanism of a combination of RANKL and CTLA4 blockade was examined by tumor-infiltrating lymphocyte analysis, tumor growth, and metastasis using a variety of neutralizing antibodies and gene-targeted mice.Results: RANKL blockade improved the efficacy of anti-CTLA4 mAbs against solid tumors and experimental metastases, with regulatory T-cell (Treg)-depleting anti-CTLA4 mAbs of the mouse IgG2a isotype showing greatest combinatorial activity. The optimal combination depended on the presence of activating Fc receptors and lymphocytes (NK cells for metastatic disease and predominantly CD8+ T cells for subcutaneous tumor control), whereas anti-RANKL alone did not require FcR. The significantly higher T-cell infiltration into solid tumors post anti-RANKL and anti-CTLA4 was accompanied by increased T-cell effector function (cytokine polyfunctionality), and anti-RANKL activity occurred independently of Treg depletion. The majority of RANKL expression in tumors was on T cells whereas RANK-expressing cells were mostly tumor-associated macrophages (TAM), with some expression also observed on dendritic cells (DC) and myeloid-derived suppressor cells (MDSC).Conclusions: These results provide a rationale for the further investigation of RANKL-RANK interactions in tumor immunity and a basis for development of translational markers of interest in human clinical trials. Clin Cancer Res; 23(19); 5789-801. ©2017 AACR.


Subject(s)
CTLA-4 Antigen/immunology , Immunotherapy , Melanoma, Experimental/therapy , RANK Ligand/immunology , Receptor Activator of Nuclear Factor-kappa B/genetics , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antigens, CD/genetics , Antigens, CD/immunology , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/genetics , Cytokines/genetics , Dendritic Cells/immunology , Gene Expression Regulation, Neoplastic/immunology , Humans , Lymphocytes/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Mice , RANK Ligand/genetics , Receptor Activator of Nuclear Factor-kappa B/immunology , T-Lymphocytes, Regulatory/immunology
14.
Cancer Discov ; 6(12): 1382-1399, 2016 12.
Article in English | MEDLINE | ID: mdl-27663893

ABSTRACT

Immunotherapy has recently entered a renaissance phase with the approval of multiple agents for the treatment of cancer. Immunotherapy stands ready to join traditional modalities, including surgery, chemotherapy, radiation, and hormone therapy, as a pillar of cancer treatment. Although immunotherapy has begun to have success in advanced cancer treatment, its scheduling and efficacy with surgery to treat earlier stages of cancer and prevent distant metastases have not been systematically examined. Here, we have used two models of spontaneously metastatic breast cancers in mice to illustrate the significantly greater therapeutic power of neoadjuvant, compared with adjuvant, immunotherapies in the context of primary tumor resection. Elevated and sustained peripheral tumor-specific immune responses underpinned the outcome, and blood sampling of tumor-specific CD8+ T cells immediately prior to and post surgery may provide a predictor of outcome. These data now provide a strong rationale to extensively test and compare neoadjuvant immunotherapy in humans. SIGNIFICANCE: We demonstrate the significantly greater therapeutic efficacy of neoadjuvant, compared with adjuvant, immunotherapies to eradicate distant metastases following primary tumor resection. Elevated and sustained peripheral tumor-specific immune responses underpinned the outcome, and blood sampling of tumor-specific CD8+ T cells immediately prior to and post surgery may provide a predictor of outcome. Cancer Discov; 6(12); 1382-99. ©2016 AACR.See related commentary by Melero et al., p. 1312This article is highlighted in the In This Issue feature, p. 1293.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/surgery , Chemotherapy, Adjuvant/methods , Neoadjuvant Therapy/methods , Animals , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Female , Humans , Immunotherapy/methods , Mice , Neoplasm Metastasis , Treatment Outcome , Xenograft Model Antitumor Assays
15.
Cancer Res ; 76(18): 5288-301, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27503925

ABSTRACT

New combination immunotherapies are displaying both efficacy and immune-related adverse events (irAE) in humans. However, grade 3/4 irAEs occur in a high proportion, which can lead to discontinuation of treatment and can result in fatalities if not promptly treated. Prolonged T regulatory cell (Treg) depletion in tumor-bearing Foxp3-DTR mice using diphtheria toxin (DT) mirrored the spectrum of antitumor responses and severity of irAEs that can occur in ipilimumab/nivolumab-treated patients. In contrast, transient Treg depletion or anti-CTLA-4/PD-1 therapy had equivalent effects in mice, lowering the immune tolerance threshold and allowing irAEs to be more easily induced following treatment with additional immunomodulatory antibodies. Transient Treg depletion of DT in combination with anti-PD-1 or anti-TIM-3 monoclonal antibodies had a high therapeutic window compared with DT plus anti-CD137. In contrast, DT plus anti-CD137-treated mice developed severe irAEs similar to grade 3/4 clinical symptoms. These irAEs appeared because of an infiltration of activated proliferating effector T cells in the tissues producing IFNγ and TNF; however, TNF blockade decreased irAEs severity without impacting on tumor growth. Cancer Res; 76(18); 5288-301. ©2016 AACR.


Subject(s)
Disease Models, Animal , Immunotherapy/adverse effects , Lymphocyte Depletion/methods , Neoplasms, Experimental , Animals , Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/adverse effects , Cell Line, Tumor , Diphtheria Toxin , Flow Cytometry , Forkhead Transcription Factors , Immunotherapy/methods , Ipilimumab , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nivolumab , T-Lymphocytes, Regulatory
16.
Immunotherapy ; 8(5): 541-53, 2016 05.
Article in English | MEDLINE | ID: mdl-27140408

ABSTRACT

AIM: We compared fresh and in vitro expanded human Tregs for their CTLA4 splicing isoform expression. METHODS: The CD4(+)CD25(+)CD127(low/-)phenotype was used for sorting Tregs and mRNA levels were measured with relative qRT-PCR. RESULTS: In fresh Tregs the level of soluble CTLA4 (sCTLA4) was half of that of full-length CTLA4, whereas in expanded cells sCTLA4 level was tenfold lower. The most striking change took place early on: sCTLA4 expression decreased significantly when cells were simply kept in culture. CONCLUSION: In the in vitro expanded Tregs, the splicing of CTLA4 is affected. Our findings can be significant for clinical cell manufacturing. First, even minimal processing of cells may impact the functional molecules. Second, Treg expansion yields more potent CTLA4 receptor bearing cells.


Subject(s)
CTLA-4 Antigen/metabolism , Protein Isoforms/metabolism , T-Lymphocytes, Regulatory/immunology , CTLA-4 Antigen/genetics , Cell Proliferation , Cells, Cultured , Gene Expression Regulation , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-7 Receptor alpha Subunit/metabolism , Protein Isoforms/genetics , Protein Splicing
17.
J Bacteriol ; 194(21): 6004, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23045508

ABSTRACT

We report the complete and annotated genome sequence of the plant-pathogenic enterobacterium Pectobacterium sp. strain SCC3193, a model strain isolated from potato in Finland. The Pectobacterium sp. SCC3193 genome consists of a 5,164,411-bp [corrected] chromosome, with no plasmids.


Subject(s)
DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Genome, Bacterial , Pectobacterium/genetics , Sequence Analysis, DNA , Finland , Molecular Sequence Data , Pectobacterium/isolation & purification , Plant Diseases/microbiology , Solanum tuberosum/microbiology
18.
Mol Plant Microbe Interact ; 21(7): 967-78, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18533837

ABSTRACT

Quorum sensing (QS), a population-density-sensing mechanism, controls the production of the main virulence determinants, the plant cell-wall-degrading enzymes (PCWDEs) of the soft-rot phytopathogen Erwinia carotovora subsp. carotovora. In this study, we used random transposon mutagenesis with a gusA reporter construct to identify two new QS-controlled genes encoding the regulator Hor and a plant ferredoxin-like protein, FerE. The QS control of the identified genes was executed by the QS regulators ExpR1 and ExpR2 and mediated by the global repressor RsmA. Hor was shown to contribute to bacterial virulence at least partly through its control of PCWDE production. Our results showed that FerE contributes to oxidative stress tolerance and in planta fitness of the bacteria and suggest that QS could be central to control of oxidative stress tolerance. The presence of the FerE protein appears to be rather unique in heterotrophic bacteria and suggests an acquisition of the corresponding gene from plant host by horizontal gene transfer.


Subject(s)
Bacterial Proteins/physiology , Host-Pathogen Interactions/physiology , Pectobacterium carotovorum/physiology , Pectobacterium carotovorum/pathogenicity , Plant Proteins/physiology , Amino Acid Sequence , Arabidopsis/genetics , Arabidopsis/microbiology , Arabidopsis/physiology , Bacterial Proteins/genetics , Base Sequence , DNA Primers/genetics , DNA, Bacterial/genetics , DNA, Plant/genetics , Genes, Bacterial , Genes, Plant , Host-Pathogen Interactions/genetics , Models, Biological , Molecular Sequence Data , Mutagenesis, Insertional , Oxidative Stress , Pectobacterium carotovorum/genetics , Plant Diseases/microbiology , Plant Proteins/genetics , Plants, Genetically Modified , Quorum Sensing/genetics , Quorum Sensing/physiology , Sequence Homology, Amino Acid , Solanum tuberosum/genetics , Solanum tuberosum/microbiology , Solanum tuberosum/physiology , Trans-Activators/genetics , Trans-Activators/physiology , Virulence/genetics , Virulence/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...