Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 16(754): eadi6887, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38959328

ABSTRACT

Virulent infectious agents such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and methicillin-resistant Staphylococcus aureus (MRSA) induce tissue damage that recruits neutrophils, monocyte, and macrophages, leading to T cell exhaustion, fibrosis, vascular leak, epithelial cell depletion, and fatal organ damage. Neutrophils, monocytes, and macrophages recruited to pathogen-infected lungs, including SARS-CoV-2-infected lungs, express phosphatidylinositol 3-kinase gamma (PI3Kγ), a signaling protein that coordinates both granulocyte and monocyte trafficking to diseased tissues and immune-suppressive, profibrotic transcription in myeloid cells. PI3Kγ deletion and inhibition with the clinical PI3Kγ inhibitor eganelisib promoted survival in models of infectious diseases, including SARS-CoV-2 and MRSA, by suppressing inflammation, vascular leak, organ damage, and cytokine storm. These results demonstrate essential roles for PI3Kγ in inflammatory lung disease and support the potential use of PI3Kγ inhibitors to suppress inflammation in severe infectious diseases.


Subject(s)
COVID-19 , Class Ib Phosphatidylinositol 3-Kinase , Inflammation , SARS-CoV-2 , COVID-19/pathology , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Animals , Inflammation/pathology , Humans , COVID-19 Drug Treatment , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice , Lung/pathology , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Cytokine Release Syndrome/drug therapy , Capillary Permeability/drug effects , Mice, Inbred C57BL , Staphylococcal Infections/drug therapy , Staphylococcal Infections/pathology
2.
Science ; 384(6702): eade8520, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38900864

ABSTRACT

Unleashing antitumor T cell activity by checkpoint inhibitor immunotherapy is effective in cancer patients, but clinical responses are limited. Cytokine signaling through the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway correlates with checkpoint immunotherapy resistance. We report a phase I clinical trial of the JAK inhibitor ruxolitinib with anti-PD-1 antibody nivolumab in Hodgkin lymphoma patients relapsed or refractory following checkpoint inhibitor immunotherapy. The combination yielded a best overall response rate of 53% (10/19). Ruxolitinib significantly reduced neutrophil-to-lymphocyte ratios and percentages of myeloid suppressor cells but increased numbers of cytokine-producing T cells. Ruxolitinib rescued the function of exhausted T cells and enhanced the efficacy of immune checkpoint blockade in preclinical solid tumor and lymphoma models. This synergy was characterized by a switch from suppressive to immunostimulatory myeloid cells, which enhanced T cell division.


Subject(s)
Hodgkin Disease , Immune Checkpoint Inhibitors , Janus Kinase Inhibitors , Nitriles , Nivolumab , Pyrazoles , Pyrimidines , T-Lymphocytes , Adult , Aged , Animals , Female , Humans , Male , Mice , Middle Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Synergism , Hodgkin Disease/drug therapy , Hodgkin Disease/immunology , Hodgkin Disease/therapy , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy , Janus Kinase Inhibitors/therapeutic use , Janus Kinases/metabolism , Janus Kinases/antagonists & inhibitors , Nitriles/therapeutic use , Nivolumab/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , T-Lymphocytes/immunology , Mice, Inbred C57BL , Mice, Inbred BALB C
3.
J Exp Clin Cancer Res ; 43(1): 157, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824552

ABSTRACT

Phosphoinositide-3-kinase γ (PI3Kγ) plays a critical role in pancreatic ductal adenocarcinoma (PDA) by driving the recruitment of myeloid-derived suppressor cells (MDSC) into tumor tissues, leading to tumor growth and metastasis. MDSC also impair the efficacy of immunotherapy. In this study we verify the hypothesis that MDSC targeting, via PI3Kγ inhibition, synergizes with α-enolase (ENO1) DNA vaccination in counteracting tumor growth.Mice that received ENO1 vaccination followed by PI3Kγ inhibition had significantly smaller tumors compared to those treated with ENO1 alone or the control group, and correlated with i) increased circulating anti-ENO1 specific IgG and IFNγ secretion by T cells, ii) increased tumor infiltration of CD8+ T cells and M1-like macrophages, as well as up-modulation of T cell activation and M1-like related transcripts, iii) decreased infiltration of Treg FoxP3+ T cells, endothelial cells and pericytes, and down-modulation of the stromal compartment and T cell exhaustion gene transcription, iv) reduction of mature and neo-formed vessels, v) increased follicular helper T cell activation and vi) increased "antigen spreading", as many other tumor-associated antigens were recognized by IgG2c "cytotoxic" antibodies. PDA mouse models genetically devoid of PI3Kγ showed an increased survival and a pattern of transcripts in the tumor area similar to that of pharmacologically-inhibited PI3Kγ-proficient mice. Notably, tumor reduction was abrogated in ENO1 + PI3Kγ inhibition-treated mice in which B cells were depleted.These data highlight a novel role of PI3Kγ in B cell-dependent immunity, suggesting that PI3Kγ depletion strengthens the anti-tumor response elicited by the ENO1 DNA vaccine.


Subject(s)
Vaccines, DNA , Animals , Mice , Vaccines, DNA/pharmacology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Humans , Cell Line, Tumor , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Disease Models, Animal , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism
4.
Blood ; 143(19): 1965-1979, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38271660

ABSTRACT

ABSTRACT: Acute myeloid leukemia (AML) is an aggressive hematological malignancy originating from transformed hematopoietic stem or progenitor cells. AML prognosis remains poor owing to resistance and relapse driven by leukemia stem cells (LSCs). Targeting molecules essential for LSC function is a promising therapeutic approach. The phosphatidylinositol 3-kinase (PI3K)/AKT pathway is often dysregulated in AML. We found that although PI3Kγ is highly enriched in LSCs and critical for self-renewal, it was dispensable for normal hematopoietic stem cells. Mechanistically, PI3Kγ-AKT signaling promotes nuclear factor erythroid 2-related factor 2 (NRF2) nuclear accumulation, which induces 6-phosphogluconate dehydrogenase (PGD) and the pentose phosphate pathway, thereby maintaining LSC stemness. Importantly, genetic or pharmacological inhibition of PI3Kγ impaired expansion and stemness of murine and human AML cells in vitro and in vivo. Together, our findings reveal a key role for PI3Kγ in selectively maintaining LSC function by regulating AKT-NRF2-PGD metabolic pathway. Targeting the PI3Kγ pathway may, therefore, eliminate LSCs without damaging normal hematopoiesis, providing a promising therapeutic strategy for AML.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase , Leukemia, Myeloid, Acute , Neoplastic Stem Cells , Pentose Phosphate Pathway , Animals , Humans , Mice , Cell Self Renewal , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Class Ib Phosphatidylinositol 3-Kinase/genetics , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Pentose Phosphate Pathway/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction
5.
Nature ; 617(7959): 42-43, 2023 05.
Article in English | MEDLINE | ID: mdl-37076711

Subject(s)
Fires , Neoplasms , Humans
6.
Nat Commun ; 13(1): 1768, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35365657

ABSTRACT

Myeloid cells play key roles in cancer immune suppression and tumor progression. In response to tumor derived factors, circulating monocytes and granulocytes extravasate into the tumor parenchyma where they stimulate angiogenesis, immune suppression and tumor progression. Chemokines, cytokines and interleukins stimulate PI3Kγ-mediated Rap1 activation, leading to conformational changes in integrin α4ß1 that promote myeloid cell extravasation and tumor inflammation Here we show that PI3Kγ activates a high molecular weight form of myosin light chain kinase, MLCK210, that promotes myosin-dependent Rap1 GTP loading, leading to integrin α4ß1 activation. Genetic or pharmacological inhibition of MLCK210 suppresses integrin α4ß1 activation, as well as tumor inflammation and progression. These results demonstrate a critical role for myeloid cell MLCK210 in tumor inflammation and serve as basis for the development of alternative approaches to develop immune oncology therapeutics.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/metabolism , Myosin-Light-Chain Kinase , Neoplasms , Cell Adhesion/physiology , Humans , Inflammation , Molecular Weight , Myeloid Cells/metabolism , Myosin-Light-Chain Kinase/metabolism , Neoplasms/genetics
7.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Article in English | MEDLINE | ID: mdl-33846242

ABSTRACT

Precision medicine in oncology leverages clinical observations of exceptional response. Toward an understanding of the molecular features that define this response, we applied an integrated, multiplatform analysis of RNA profiles derived from clinically annotated glioblastoma samples. This analysis suggested that specimens from exceptional responders are characterized by decreased accumulation of microglia/macrophages in the glioblastoma microenvironment. Glioblastoma-associated microglia/macrophages secreted interleukin 11 (IL11) to activate STAT3-MYC signaling in glioblastoma cells. This signaling induced stem cell states that confer enhanced tumorigenicity and resistance to the standard-of-care chemotherapy, temozolomide (TMZ). Targeting a myeloid cell restricted an isoform of phosphoinositide-3-kinase, phosphoinositide-3-kinase gamma isoform (PI3Kγ), by pharmacologic inhibition or genetic inactivation disrupted this signaling axis by reducing microglia/macrophage-associated IL11 secretion in the tumor microenvironment. Mirroring the clinical outcomes of exceptional responders, PI3Kγ inhibition synergistically enhanced the anti-neoplastic effects of TMZ in orthotopic murine glioblastoma models. Moreover, inhibition or genetic inactivation of PI3Kγ in murine glioblastoma models recapitulated expression profiles observed in clinical specimens isolated from exceptional responders. Our results suggest key contributions from tumor-associated microglia/macrophages in exceptional responses and highlight the translational potential for PI3Kγ inhibition as a glioblastoma therapy.


Subject(s)
Glioblastoma/metabolism , Microglia/metabolism , Temozolomide/pharmacology , Adult , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Drug Resistance, Neoplasm/physiology , Female , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Interleukin-11/immunology , Interleukin-11/metabolism , Male , Mice, Inbred C57BL , Mice, Nude , Microglia/physiology , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Temozolomide/metabolism , Tumor Microenvironment/drug effects , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/physiology
8.
Cancer Res ; 79(19): 5048-5059, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31416839

ABSTRACT

Tumor-associated macrophages (TAM) are highly expressed within the tumor microenvironment of a wide range of cancers, where they exert a protumor phenotype by promoting tumor cell growth and suppressing antitumor immune function. Here, we show that TAM accumulation in human and mouse tumors correlates with tumor cell expression of integrin αvß3, a known driver of epithelial cancer progression and drug resistance. A monoclonal antibody targeting αvß3 (LM609) exploited the coenrichment of αvß3 and TAMs to not only eradicate highly aggressive drug-resistant human lung and pancreas cancers in mice, but also to prevent the emergence of circulating tumor cells. Importantly, this antitumor activity in mice was eliminated following macrophage depletion. Although LM609 had no direct effect on tumor cell viability, it engaged macrophages but not natural killer (NK) cells to induce antibody-dependent cellular cytotoxicity (ADCC) of αvß3-expressing tumor cells despite their expression of the CD47 "don't eat me" signal. In contrast to strategies designed to eliminate TAMs, these findings suggest that anti-αvß3 represents a promising immunotherapeutic approach to redirect TAMs to serve as tumor killers for late-stage or drug-resistant cancers. SIGNIFICANCE: Therapeutic antibodies are commonly engineered to optimize engagement of NK cells as effectors. In contrast, LM609 targets αvß3 to suppress tumor progression and enhance drug sensitivity by exploiting TAMs to trigger ADCC.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity/drug effects , Integrin alphaVbeta3/immunology , Macrophages/immunology , Neoplasms, Glandular and Epithelial/immunology , Animals , Antineoplastic Agents/pharmacology , Disease Progression , Humans , Mice , Neoplasms, Glandular and Epithelial/pathology , Phagocytosis/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
9.
Adv Mater ; 31(35): e1902952, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31267590

ABSTRACT

Despite the promise of ribonucleic acid interference therapeutics, the delivery of oligonucleotides selectively to diseased tissues in the body, and specifically to the cellular location in the tissues needed to provide optimal therapeutic outcome, remains a significant challenge. Here, key material properties and biological mechanisms for delivery of short interfering RNAs (siRNAs) to effectively silence target-specific cells in vivo are identified. Using porous silicon nanoparticles as the siRNA host, tumor-targeting peptides for selective tissue homing, and fusogenic lipid coatings to induce fusion with the plasma membrane, it is shown that the uptake mechanism can be engineered to be independent of common receptor-mediated endocytosis pathways. Two examples of the potential broad clinical applicability of this concept in a mouse xenograft model of ovarian cancer peritoneal carcinomatosis are provided: silencing the Rev3l subunit of polymerase Pol ζ to impair DNA repair in combination with cisplatin; and reprogramming tumor-associated macrophages into a proinflammatory state.


Subject(s)
Drug Carriers/chemistry , Drug Carriers/metabolism , Endosomes/metabolism , Nanoparticles/chemistry , Peptides/metabolism , RNA, Small Interfering/chemistry , Silicon/chemistry , Animals , Cell Line, Tumor , Gene Silencing , Humans , Mice , Porosity , RNA, Small Interfering/genetics
10.
Oncogene ; 38(28): 5599-5611, 2019 07.
Article in English | MEDLINE | ID: mdl-30967626

ABSTRACT

The MST1R (RON) kinase is overexpressed in >80% of human pancreatic cancers, but its role in pancreatic carcinogenesis is unknown. In this study, we examined the relevance of Mst1r kinase to Kras driven pancreatic carcinogenesis using genetically engineered mouse models. In the setting of mutant Kras, Mst1r overexpression increased acinar-ductal metaplasia (ADM), accelerated the progression of pancreatic intraepithelial neoplasia (PanIN), and resulted in the accumulation of (mannose receptor C type 1) MRC1+, (arginase 1) Arg+ macrophages in the tumor microenvironment. Conversely, absence of a functional Mst1r kinase slowed PanIN initiation, resulted in smaller tumors, prolonged survival and a reduced tumor-associated macrophage content. Mst1r expression was associated with increased production of its ligand Mst1, and in orthotopic models, suppression of Mst1 expression resulted in reduced tumor size, changes in macrophage polarization and enhanced T cell infiltration. This study demonstrates the functional significance of Mst1r during pancreatic cancer initiation and progression. Further, it provides proof of concept that targeting Mst1r can modulate pancreatic cancer growth and the microenvironment. This study provides further rationale for targeting Mst1r as a therapeutic strategy.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Epithelial Cells/pathology , Macrophages/pathology , Pancreatic Neoplasms/pathology , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Carcinoma, Pancreatic Ductal/enzymology , Disease Progression , Female , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pancreatic Neoplasms/enzymology , Proof of Concept Study , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction , Tumor Microenvironment
11.
Trends Immunol ; 40(4): 310-327, 2019 04.
Article in English | MEDLINE | ID: mdl-30890304

ABSTRACT

Macrophages are phagocytes that serve as a first line of defense against pathogenic insults to tissues. These innate immune cells mount proinflammatory responses to pathogens and repair damaged tissues. However, tumor-associated macrophages (TAMs) express cytokines and chemokines that can suppress antitumor immunity and promote tumor progression. Preclinical studies have identified crucial pathways regulating the recruitment, polarization, and metabolism of TAMs during tumor progression. Moreover, novel therapeutics targeting these pathways can indirectly stimulate cytotoxic T cell activation and recruitment, and synergize with checkpoint inhibitors, chemotherapy and/or radiation therapy in preclinical studies. Thus, clinical trials with therapeutic agents that promote phagocytosis or suppress survival, proliferation, trafficking, or polarization of TAMs are currently underway. These early results offer the promise of improved cancer outcomes.


Subject(s)
Macrophages/immunology , Neoplasms/immunology , Animals , Humans , Neoplasms/therapy
12.
Nat Commun ; 9(1): 5379, 2018 12 19.
Article in English | MEDLINE | ID: mdl-30568188

ABSTRACT

Myeloid cells are recruited to damaged tissues where they can resolve infections and tumor growth or stimulate wound healing and tumor progression. Recruitment of these cells is regulated by integrins, a family of adhesion receptors that includes integrin CD11b. Here we report that, unexpectedly, integrin CD11b does not regulate myeloid cell recruitment to tumors but instead controls myeloid cell polarization and tumor growth. CD11b activation promotes pro-inflammatory macrophage polarization by stimulating expression of microRNA Let7a. In contrast, inhibition of CD11b prevents Let7a expression and induces cMyc expression, leading to immune suppressive macrophage polarization, vascular maturation, and accelerated tumor growth. Pharmacological activation of CD11b with a small molecule agonist, Leukadherin 1 (LA1), promotes pro-inflammatory macrophage polarization and suppresses tumor growth in animal models of murine and human cancer. These studies identify CD11b as negative regulator of immune suppression and a target for cancer immune therapy.


Subject(s)
Benzoates/therapeutic use , CD11b Antigen/metabolism , Macrophages/metabolism , Melanoma, Experimental/immunology , MicroRNAs/metabolism , Thiohydantoins/therapeutic use , Animals , Benzoates/pharmacology , CD11b Antigen/agonists , Macrophages/drug effects , Melanoma, Experimental/drug therapy , Mice, Transgenic , Neovascularization, Pathologic , Proto-Oncogene Proteins c-myc/metabolism , Thiohydantoins/pharmacology
13.
Cancer Immunol Res ; 5(11): 957-968, 2017 11.
Article in English | MEDLINE | ID: mdl-28963139

ABSTRACT

Immunosuppressive myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) accumulate in tumors where they inhibit T cell-mediated antitumor immune responses and promote tumor progression. Myeloid cell PI3Kγ plays a role in regulating tumor immune suppression by promoting integrin α4-dependent MDSC recruitment to tumors and by stimulating the immunosuppressive polarization of MDSCs and TAMs. Here, we show that integrin α4 promotes immunosuppressive polarization of MDSCs and TAMs downstream of PI3Kγ, thereby inhibiting antitumor immunity. Genetic or pharmacological suppression of either PI3Kγ or integrin α4 blocked MDSC recruitment to tumors and also inhibited immune suppressive myeloid cell polarization, thereby reducing expression of IL10 and increasing expression of IL12 and IFNγ within tumors. Inhibition of PI3Kγ or integrin α4 within tumors stimulated dendritic cell and CD8+ T-cell recruitment and maturation, as well as tumor cell cytotoxicity in vivo, thereby inhibiting tumor growth. As blockade of PI3Kγ or integrin α4 prevents accumulation of MDSC and reduces myeloid cell expression of immunosuppressive factors that stimulate tumor immune escape, these results highlight PI3Kγ and integrin α4 as targets for the design of cancer therapeutics. Cancer Immunol Res; 5(11); 957-68. ©2017 AACR.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/immunology , Integrin alpha4beta1/immunology , Neoplasms/immunology , Animals , Antibodies/pharmacology , Cell Line, Tumor , Cell Polarity/drug effects , Cells, Cultured , Class Ib Phosphatidylinositol 3-Kinase/genetics , Cytokines/immunology , Female , Immune Tolerance , Integrin alpha4beta1/antagonists & inhibitors , Lymphocytes, Tumor-Infiltrating/immunology , Macrophages/immunology , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/drug effects , Myeloid Cells/immunology , Neoplasms/drug therapy , Neoplasms/pathology , Phosphoinositide-3 Kinase Inhibitors
14.
JCI Insight ; 2(18)2017 09 21.
Article in English | MEDLINE | ID: mdl-28931759

ABSTRACT

Checkpoint inhibitors have demonstrated efficacy in patients with recurrent or metastatic head and neck squamous cell carcinoma (HNSCC). However, the majority of patients do not benefit from these agents. To improve the efficacy of checkpoint inhibitors, intratumoral (i.t.) injection with innate immune activators, TLR7 and TLR9 agonists, were tested along with programmed death-1 receptor (PD-1) blockade. The combination therapy suppressed tumor growth at the primary injected and distant sites in human papillomavirus-negative (HPV-negative) SCC7 and MOC1, and HPV-positive MEER syngeneic mouse models. Abscopal effects and suppression of secondary challenged tumor suggest that local treatment with TLR agonists in combination with anti-PD-1 provided systemic adaptive immunity. I.t. treatment with a TLR7 agonist increased the ratio of M1 to M2 tumor-associated macrophages (TAMs) and promoted the infiltration of tumor-specific IFNγ-producing CD8+ T cells. Anti-PD-1 treatment increased T cell receptor (TCR) clonality of CD8+ T cells in tumors and spleens of treated mice. Collectively, these experiments demonstrate that combination therapy with i.t. delivery of TLR agonists and PD-1 blockade activates TAMs and induces tumor-specific adaptive immune responses, leading to suppression of primary tumor growth and prevention of metastasis in HNSCC models.


Subject(s)
Head and Neck Neoplasms/therapy , Immunotherapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Squamous Cell Carcinoma of Head and Neck/therapy , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Heterografts , Humans , Killer Cells, Natural/immunology , Macrophages/immunology , Mice , Mice, Inbred C3H , Toll-Like Receptor 7/agonists , Toll-Like Receptor 9/agonists , Tumor Microenvironment
16.
Nature ; 539(7629): 437-442, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27642729

ABSTRACT

Macrophages play critical, but opposite, roles in acute and chronic inflammation and cancer. In response to pathogens or injury, inflammatory macrophages express cytokines that stimulate cytotoxic T cells, whereas macrophages in neoplastic and parasitic diseases express anti-inflammatory cytokines that induce immune suppression and may promote resistance to T cell checkpoint inhibitors. Here we show that macrophage PI 3-kinase γ controls a critical switch between immune stimulation and suppression during inflammation and cancer. PI3Kγ signalling through Akt and mTor inhibits NFκB activation while stimulating C/EBPß activation, thereby inducing a transcriptional program that promotes immune suppression during inflammation and tumour growth. By contrast, selective inactivation of macrophage PI3Kγ stimulates and prolongs NFκB activation and inhibits C/EBPß activation, thus promoting an immunostimulatory transcriptional program that restores CD8+ T cell activation and cytotoxicity. PI3Kγ synergizes with checkpoint inhibitor therapy to promote tumour regression and increased survival in mouse models of cancer. In addition, PI3Kγ-directed, anti-inflammatory gene expression can predict survival probability in cancer patients. Our work thus demonstrates that therapeutic targeting of intracellular signalling pathways that regulate the switch between macrophage polarization states can control immune suppression in cancer and other disorders.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/metabolism , Immune Tolerance/immunology , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cells, Cultured , Class Ib Phosphatidylinositol 3-Kinase/deficiency , Class Ib Phosphatidylinositol 3-Kinase/genetics , Female , Humans , Inflammation/immunology , Lymphocyte Activation , Macrophages/enzymology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Neoplasms/immunology , Neoplasms/pathology , Phosphoinositide-3 Kinase Inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/immunology , TOR Serine-Threonine Kinases/metabolism , Tumor Escape/immunology
17.
Cancer Discov ; 6(8): 870-85, 2016 08.
Article in English | MEDLINE | ID: mdl-27179037

ABSTRACT

UNLABELLED: Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with a low 5-year survival rate, yet new immunotherapeutic modalities may offer hope for this and other intractable cancers. Here, we report that inhibitory targeting of PI3Kγ, a key macrophage lipid kinase, stimulates antitumor immune responses, leading to improved survival and responsiveness to standard-of-care chemotherapy in animal models of PDAC. PI3Kγ selectively drives immunosuppressive transcriptional programming in macrophages that inhibits adaptive immune responses and promotes tumor cell invasion and desmoplasia in PDAC. Blockade of PI3Kγ in PDAC-bearing mice reprograms tumor-associated macrophages to stimulate CD8(+) T-cell-mediated tumor suppression and to inhibit tumor cell invasion, metastasis, and desmoplasia. These data indicate the central role that macrophage PI3Kγ plays in PDAC progression and demonstrate that pharmacologic inhibition of PI3Kγ represents a new therapeutic modality for this devastating tumor type. SIGNIFICANCE: We report here that PI3Kγ regulates macrophage transcriptional programming, leading to T-cell suppression, desmoplasia, and metastasis in pancreas adenocarcinoma. Genetic or pharmacologic inhibition of PI3Kγ restores antitumor immune responses and improves responsiveness to standard-of-care chemotherapy. PI3Kγ represents a new therapeutic immune target for pancreas cancer. Cancer Discov; 6(8); 870-85. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 803.


Subject(s)
Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Macrophages/metabolism , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Biomarkers , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement/genetics , Class Ib Phosphatidylinositol 3-Kinase/genetics , Disease Models, Animal , Disease Progression , Gene Expression , Gene Knockout Techniques , Heterografts , Humans , Immunomodulation , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages/immunology , Male , Mice , Mice, Knockout , Mice, Transgenic , Mortality , Neoplasm Metastasis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phenols/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Pteridines/pharmacology , Xenograft Model Antitumor Assays
18.
Cancer Discov ; 6(3): 270-85, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26715645

ABSTRACT

UNLABELLED: Pancreas ductal adenocarcinoma (PDAC) has one of the worst 5-year survival rates of all solid tumors, and thus new treatment strategies are urgently needed. Here, we report that targeting Bruton tyrosine kinase (BTK), a key B-cell and macrophage kinase, restores T cell-dependent antitumor immune responses, thereby inhibiting PDAC growth and improving responsiveness to standard-of-care chemotherapy. We report that PDAC tumor growth depends on cross-talk between B cells and FcRγ(+) tumor-associated macrophages, resulting in T(H)2-type macrophage programming via BTK activation in a PI3Kγ-dependent manner. Treatment of PDAC-bearing mice with the BTK inhibitor PCI32765 (ibrutinib) or by PI3Kγ inhibition reprogrammed macrophages toward a T(H)1 phenotype that fostered CD8(+) T-cell cytotoxicity, and suppressed PDAC growth, indicating that BTK signaling mediates PDAC immunosuppression. These data indicate that pharmacologic inhibition of BTK in PDAC can reactivate adaptive immune responses, presenting a new therapeutic modality for this devastating tumor type. SIGNIFICANCE: We report that BTK regulates B-cell and macrophage-mediated T-cell suppression in pancreas adenocarcinomas. Inhibition of BTK with the FDA-approved inhibitor ibrutinib restores T cell-dependent antitumor immune responses to inhibit PDAC growth and improves responsiveness to chemotherapy, presenting a new therapeutic modality for pancreas cancer.


Subject(s)
Cell Communication/immunology , Immune System/cytology , Immune System/pathology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Protein-Tyrosine Kinases/metabolism , Agammaglobulinaemia Tyrosine Kinase , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Disease Models, Animal , Disease Progression , Humans , Leukocytes/immunology , Leukocytes/metabolism , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Pancreatic Neoplasms/genetics , Receptors, IgG/metabolism , Signal Transduction
19.
PLoS One ; 10(5): e0120815, 2015.
Article in English | MEDLINE | ID: mdl-25973900

ABSTRACT

High-risk neuroblastoma is associated with an overall survival rate of 30-50%. Neuroblastoma-expressed cell adhesion receptors of the integrin family impact cell adhesion, migration, proliferation and survival. Integrin α4 is essential for neural crest cell motility during development, is highly expressed on leukocytes, and is critical for transendothelial migration. Thus, cancer cells that express this receptor may exhibit increased metastatic potential. We show that α4 expression in human and murine neuroblastoma cell lines selectively enhances in vitro interaction with the alternatively spliced connecting segment 1 of fibronectin, as well as vascular cell adhesion molecule-1 and increases migration. Integrin α4 expression enhanced experimental metastasis in a syngeneic tumor model, reconstituting a pattern of organ involvement similar to that seen in patients. Accordingly, antagonism of integrin α4 blocked metastasis, suggesting adhesive function of the integrin is required. However, adhesive function was not sufficient, as mutants of integrin α4 that conserved the matrix-adhesive and promigratory function in vitro were compromised in their metastatic capacity in vivo. Clinically, integrin α4 is more frequently expressed in non-MYNC amplified tumors, and is selectively associated with poor prognosis in this subset of disease. These results reveal an unexpected role for integrin α4 in neuroblastoma dissemination and identify α4 as a potential prognostic indicator and therapeutic target.


Subject(s)
Gene Expression Regulation, Neoplastic , Integrin alpha4/genetics , Liver Neoplasms/genetics , Nervous System Neoplasms/genetics , Neuroblastoma/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Animals , Cell Adhesion , Cell Line, Tumor , Cell Movement , Humans , Integrin alpha4/metabolism , Intercellular Signaling Peptides and Proteins , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Lymphatic Metastasis , Mice , N-Myc Proto-Oncogene Protein , Neoplasm Transplantation , Nervous System Neoplasms/metabolism , Nervous System Neoplasms/pathology , Neuroblastoma/metabolism , Neuroblastoma/secondary , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Peptides/genetics , Peptides/metabolism , Prognosis , Signal Transduction , Survival Analysis , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
20.
PLoS One ; 8(4): e60226, 2013.
Article in English | MEDLINE | ID: mdl-23565202

ABSTRACT

Tumor inflammation, the recruitment of myeloid lineage cells into the tumor microenvironment, promotes angiogenesis, immunosuppression and metastasis. CD11b+Gr1lo monocytic lineage cells and CD11b+Gr1hi granulocytic lineage cells are recruited from the circulation by tumor-derived chemoattractants, which stimulate PI3-kinase γ (PI3Kγ)-mediated integrin α4 activation and extravasation. We show here that PI3Kγ activates PLCγ, leading to RasGrp/CalDAG-GEF-I&II mediated, Rap1a-dependent activation of integrin α4ß1, extravasation of monocytes and granulocytes, and inflammation-associated tumor progression. Genetic depletion of PLCγ, CalDAG-GEFI or II, Rap1a, or the Rap1 effector RIAM was sufficient to prevent integrin α4 activation by chemoattractants or activated PI3Kγ (p110γCAAX), while activated Rap (RapV12) promoted constitutive integrin activation and cell adhesion that could only be blocked by inhibition of RIAM or integrin α4ß1. Similar to blockade of PI3Kγ or integrin α4ß1, blockade of Rap1a suppressed both the recruitment of monocytes and granulocytes to tumors and tumor progression. These results demonstrate critical roles for a PI3Kγ-Rap1a-dependent pathway in integrin activation during tumor inflammation and suggest novel avenues for cancer therapy.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/metabolism , Inflammation/metabolism , Integrin alpha4beta1/metabolism , Myeloid Cells/metabolism , Neoplasms/metabolism , rap1 GTP-Binding Proteins/metabolism , Animals , Female , Guanine Nucleotide Exchange Factors/metabolism , Humans , Inflammation/genetics , Integrin alpha4beta1/chemistry , Ligands , Mice , Mice, Knockout , Neoplasms/genetics , Phospholipase C gamma/metabolism , Protein Binding , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...