Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Immunol Rev ; 314(1): 13-35, 2023 03.
Article in English | MEDLINE | ID: mdl-36527200

ABSTRACT

Neutrophils sense microbes and host inflammatory mediators, and traffic to sites of infection where they direct a broad armamentarium of antimicrobial products against pathogens. Neutrophils are also activated by damage-associated molecular patterns (DAMPs), which are products of cellular injury that stimulate the innate immune system through pathways that are similar to those activated by microbes. Neutrophils and platelets become activated by injury, and cluster and cross-signal to each other with the cumulative effect of driving antimicrobial defense and hemostasis. In addition, neutrophil extracellular traps are extracellular chromatin and granular constituents that are generated in response to microbial and damage motifs and are pro-thrombotic and injurious. Although neutrophils can worsen tissue injury, neutrophils may also have a role in facilitating wound repair following injury. A central theme of this review relates to how critical functions of neutrophils that evolved to respond to infection and damage modulate the tumor microenvironment (TME) in ways that can promote or limit tumor progression. Neutrophils are reprogrammed by the TME, and, in turn, can cross-signal to tumor cells and reshape the immune landscape of tumors. Importantly, promising new therapeutic strategies have been developed to target neutrophil recruitment and function to make cancer immunotherapy more effective.


Subject(s)
Extracellular Traps , Neutrophils , Humans , Blood Platelets/metabolism , Blood Platelets/pathology , Endothelial Cells , Inflammation , T-Lymphocytes , Extracellular Traps/metabolism
2.
Cancer Immunol Immunother ; 71(10): 2355-2369, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35166871

ABSTRACT

The ovarian tumor microenvironment (TME) is characterized by the accumulation of immunosuppressive tumor-associated macrophages (TAMs) and granulocytic cells. Very small size particles (VSSP), comprised of the ganglioside NAcGM3 and Neisseria meningitidis derived outer membrane vesicles, is being developed as a nanoparticulated modulator of innate immunity. Prior studies have shown that VSSP enhanced antigen-specific cytotoxic T cell responses and reduced the suppressive phenotype of splenic granulocytic cells in tumor-bearing mice. Here, we hypothesized that intraperitoneal VSSP would modify myeloid cell accumulation and phenotypes in the ovarian TME and abrogate suppressor function of TAMs and tumor-associated granulocytic cells. In the ID8 syngeneic model of epithelial ovarian cancer, VSSP reduced peritoneal TAMs and induced M1-like polarization in TAMs. In addition, VSSP stimulated peritoneal inflammation characterized by increased granulocytes and monocytes, including inflammatory monocytic cells. VSSP treatment resulted in peritoneal TAMs and granulocytic cells being less suppressive of ex vivo stimulated CD8+ T cell responses. VSSP alone and combined with anti-PD-1 modestly but significantly prolonged survival in tumor-bearing mice. In addition, ex vivo treatment with VSSP induced M1-like polarization in TAMs from patients with metastatic ovarian cancer and variably abrogated their suppressor phenotype. VSSP treatment also partially abrogated the induction of suppressor function in healthy donor neutrophils exposed to ascites supernatants from patients with ovarian cancer. Together, these results point to VSSP reprogramming myeloid responses resulting in abrogation of suppressive pathways and raise the potential for administration of VSSP into the TME to enhance anti-tumor immunity.


Subject(s)
Ovarian Neoplasms , Tumor-Associated Macrophages , Animals , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred C57BL , Myeloid Cells , Tumor Microenvironment
3.
Cancer Immunol Res ; 9(7): 790-810, 2021 07.
Article in English | MEDLINE | ID: mdl-33990375

ABSTRACT

T-cell activation and expansion in the tumor microenvironment (TME) are critical for antitumor immunity. Neutrophils in the TME acquire a complement-dependent T-cell suppressor phenotype that is characterized by inhibition of T-cell proliferation and activation through mechanisms distinct from those of myeloid-derived suppressor cells. In this study, we used ascites fluid supernatants (ASC) from patients with ovarian cancer as an authentic component of the TME to evaluate the effects of ASC on neutrophil function and mechanisms for neutrophil-driven immune suppression. ASC prolonged neutrophil life span, decreased neutrophil density, and induced nuclear hypersegmentation. Mass cytometry analysis showed that ASC induced 15 distinct neutrophil clusters. ASC stimulated complement deposition and signaling in neutrophils, resulting in surface mobilization of granule constituents, including NADPH oxidase. NADPH oxidase activation and phosphatidylserine signaling were required for neutrophil suppressor function, although we did not observe a direct role of extracellular reactive oxygen species in inhibiting T-cell proliferation. Postoperative surgical drainage fluid also induced a complement-dependent neutrophil suppressor phenotype, pointing to this effect as a general response to injury. Like circulating lymphocytes, ASC-activated neutrophils caused complement-dependent suppression of tumor-associated lymphocytes. ASC-activated neutrophils adhered to T cells and caused trogocytosis of T-cell membranes. These injury and signaling cues resulted in T-cell immunoparalysis characterized by impaired NFAT translocation, IL2 production, glucose uptake, mitochondrial function, and mTOR activation. Our results demonstrate that complement-dependent priming of neutrophil effector functions in the TME induces a T-cell nonresponsiveness distinct from established checkpoint pathways and identify targets for immunotherapy.See related Spotlight by Cassatella, p. 725.


Subject(s)
Neutrophils/immunology , Ovarian Neoplasms/immunology , T-Lymphocytes/immunology , Trogocytosis/immunology , Tumor Escape , Adult , Cells, Cultured , Female , Humans , Lymphocyte Activation , Middle Aged , Neutrophil Activation , Neutrophils/metabolism , Ovarian Neoplasms/blood , Ovarian Neoplasms/pathology , Ovarian Neoplasms/surgery , Primary Cell Culture , Tumor Microenvironment/immunology , Young Adult
4.
Cancer Immunol Res ; 8(6): 819-828, 2020 06.
Article in English | MEDLINE | ID: mdl-32238380

ABSTRACT

Myeloid derived suppressor cells (MDSC) are a heterogeneous group of immature cells that accumulate in the peripheral blood and tumor microenvironment and are barriers to cancer therapy. MDSCs serve as prognostic biomarkers and are targets for therapy. On the basis of surface markers, three subsets of MDSCs have been defined in humans: granulocytic, monocytic, and early stage (e-MDSC). The markers attributed to e-MDSCs overlap with those of basophils, which are rare circulating myeloid cells with unrecognized roles in cancer. Thus, we asked whether e-MDSCs in circulation and the tumor microenvironment include basophils. On average, 58% of cells with e-MDSC surface markers in blood and 36% in ascites from patients with ovarian cancer were basophils based on CD123high expression and cytology, whereas cells with immature features were rare. Circulating and ascites basophils did not suppress proliferation of stimulated T cells, a key feature of MDSCs. Increased accumulation of basophils and basogranulin, a marker of basophil degranulation, were observed in ascites compared to serum in patients with newly diagnosed ovarian cancer. Basophils recruited to the tumor microenvironment may exacerbate fluid accumulation by their release of proinflammatory granular constituents that promote vascular leakage. No significant correlation was observed between peripheral basophil counts and survival in patients with ovarian cancer. Our results suggest that studies in which e-MDSCs were defined solely by surface markers should be reevaluated to exclude basophils. Both immaturity and suppression are criteria to define e-MDSCs in future studies.


Subject(s)
Ascites/pathology , Basophils/pathology , Biomarkers, Tumor/blood , Leukocytes, Mononuclear/pathology , Myeloid-Derived Suppressor Cells/pathology , Ovarian Neoplasms/pathology , Tumor Microenvironment , Case-Control Studies , Female , Humans , Middle Aged , Ovarian Neoplasms/blood , Ovarian Neoplasms/surgery , Prognosis , Prospective Studies , Retrospective Studies , Survival Rate , Tumor Cells, Cultured
5.
JCI Insight ; 4(5)2019 03 07.
Article in English | MEDLINE | ID: mdl-30730851

ABSTRACT

Epithelial ovarian cancer (EOC) often presents with metastases and ascites. Granulocytic myeloid-derived suppressor cells are an immature population that impairs antitumor immunity. Since suppressive granulocytes in the ascites of patients with newly diagnosed EOC were morphologically mature, we hypothesized that PMN were rendered suppressive in the tumor microenvironment (TME). Circulating PMN from patients were not suppressive but acquired a suppressor phenotype (defined as ≥1 log10 reduction of anti-CD3/CD28-stimulated T cell proliferation) after ascites supernatant exposure. Ascites supernatants (20 of 31 supernatants) recapitulated the suppressor phenotype in PMN from healthy donors. T cell proliferation was restored with ascites removal and restimulation. PMN suppressors also inhibited T cell activation and cytokine production. PMN suppressors completely suppressed proliferation in naive, central memory, and effector memory T cells and in engineered tumor antigen-specific cytotoxic T lymphocytes, while antigen-specific cell lysis was unaffected. Inhibition of complement C3 activation and PMN effector functions, including CR3 signaling, protein synthesis, and vesicular trafficking, abrogated the PMN suppressor phenotype. Moreover, malignant effusions from patients with various metastatic cancers also induced the C3-dependent PMN suppressor phenotype. These results point to PMN impairing T cell expansion and activation in the TME and the potential for complement inhibition to abrogate this barrier to antitumor immunity.


Subject(s)
Immunity , Neutrophils/immunology , Ovarian Neoplasms/immunology , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Aged , CD28 Antigens , Cell Proliferation , Complement C3 , Cytokines , Female , Granulocytes , Humans , Lymphocyte Activation/immunology , Muromonab-CD3 , Myeloid-Derived Suppressor Cells/immunology , Ovarian Neoplasms/pathology
6.
Gynecol Oncol ; 138(2): 352-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26001328

ABSTRACT

OBJECTIVES: Epithelial ovarian cancer (EOC) typically presents with advanced disease. Even with optimal debulking and response to adjuvant chemotherapy, the majority of patients will have disease relapse. We evaluated cytokine and chemokine profiles in ascites at primary surgery as biomarkers for progression-free survival (PFS) and overall survival (OS) in patients with advanced EOC. METHODS: Retrospective analysis of patients (n =70) who underwent surgery at Roswell Park Cancer Institute between 2002 and 2012, followed by platinum-based chemotherapy. RESULTS: The mean age at diagnosis was 61.8 years, 85.3% had serous EOC, and 95.7% had stage IIIB, IIIC, or IV disease. Univariate analysis showed that ascites levels of tumor necrosis factor (TNF)-α were associated with reduced PFS after primary surgery. Although the ascites concentration of interleukin (IL)-6 was not by itself predictive of PFS, we found that stratifying patients by high TNF-α and high IL-6 levels identified a sub-group of patients at high risk for rapid disease relapse. This effect was largely independent of clinical prognostic variables. CONCLUSIONS: The combination of high TNF-α and high IL-6 ascites levels at primary surgery predicts worse PFS in patients with advanced EOC. These results suggest an interaction between ascites TNF-α and IL-6 in driving tumor progression and resistance to chemotherapy in advanced EOC, and raise the potential for pre-treatment ascites levels of these cytokines as prognostic biomarkers. This study involved a small sample of patients and was an exploratory analysis; therefore, findings require validation in a larger independent cohort.


Subject(s)
Ascites/metabolism , Interleukin-6/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adult , Age Factors , Aged , Aged, 80 and over , Ascites/immunology , Ascites/pathology , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Carcinoma, Ovarian Epithelial , Disease-Free Survival , Female , Humans , Interleukin-6/immunology , Interleukin-8/immunology , Interleukin-8/metabolism , Middle Aged , Neoplasms, Glandular and Epithelial/immunology , Neoplasms, Glandular and Epithelial/pathology , Neoplasms, Glandular and Epithelial/surgery , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/surgery , Prognosis , Survival Rate , Tumor Necrosis Factor-alpha/immunology , Young Adult
7.
Oncotarget ; 6(13): 11310-26, 2015 May 10.
Article in English | MEDLINE | ID: mdl-25888637

ABSTRACT

Epithelial ovarian cancer (EOC) is typically diagnosed at advanced stages, and is associated with a high relapse rate. Patients in remission are ideal candidates for immunotherapy aimed at cure or prolonging disease-free periods. However, immunosuppressive pathways in the tumor microenvironment are obstacles to durable anti-tumor immunity. In a metastatic syngeneic mouse model of EOC, immunosuppressive macrophages and myeloid-derived suppressor cells (MDSCs) accumulate in the local tumor environment. In addition, resident peritoneal macrophages from non-tumor-bearing mice were highly immunosuppressive, abrogating stimulated T cell proliferation in a cell contact-dependent manner. Immunization with microparticles containing TLR9 and NOD-2 ligands (MIS416) significantly prolonged survival in tumor-bearing mice. The strategy of MIS416 immunization followed by anti-CD11b administration further delayed tumor progression, thereby establishing the proof of principle that myeloid depletion can enhance vaccine efficacy. In patients with advanced EOC, ascites analysis showed substantial heterogeneity in the relative proportions of myeloid subsets and their immunosuppressive properties. Together, these findings point to immunosuppressive myeloid cells in the EOC microenvironment as targets to enhance vaccination. Further studies of myeloid cell accumulation and functional phenotypes in the EOC microenvironment may identify patients who are likely to benefit from vaccination combined with approaches that deplete tumor-associated myeloid cells.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Cancer Vaccines/administration & dosage , Myeloid Cells/immunology , Neoplasms, Glandular and Epithelial/therapy , Ovarian Neoplasms/therapy , Tumor Escape , Tumor Microenvironment , Vaccination , Adoptive Transfer , Animals , Antibodies, Monoclonal/immunology , Ascites/immunology , CD11b Antigen/immunology , Cancer Vaccines/immunology , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Disease Progression , Female , Humans , Ligands , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Macrophages/immunology , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/immunology , Neoplasms, Glandular and Epithelial/pathology , Nod2 Signaling Adaptor Protein/immunology , Ovalbumin/administration & dosage , Ovalbumin/immunology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , T-Lymphocytes/immunology , Time Factors , Toll-Like Receptor 9/immunology
SELECTION OF CITATIONS
SEARCH DETAIL