Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 45
1.
Front Immunol ; 15: 1341390, 2024.
Article En | MEDLINE | ID: mdl-38426089

The tumor microenvironment is composed of tumor cells, stromal cells and leukocytes, including innate and adaptive immune cells, and represents an ecological niche that regulates tumor development and progression. In general, inflammatory cells are considered to contribute to tumor progression through various mechanisms, including the formation of an immunosuppressive microenvironment. Macrophages and neutrophils are important components of the tumor microenvironment and can act as a double-edged sword, promoting or inhibiting the development of the tumor. Targeting of the immune system is emerging as an important therapeutic strategy for cancer patients. However, the efficacy of the various immunotherapies available is still limited. Given the crucial importance of the crosstalk between macrophages and neutrophils and other immune cells in the formation of the anti-tumor immune response, targeting these interactions may represent a promising therapeutic approach against cancer. Here we will review the current knowledge of the role played by macrophages and neutrophils in cancer, focusing on their interaction with other immune cells.


Neoplasms , Neutrophils , Humans , Macrophages , Immunity , Immunotherapy , Tumor Microenvironment
2.
Cancer Immunol Res ; 12(4): 413-426, 2024 Apr 02.
Article En | MEDLINE | ID: mdl-38349973

Neutrophils are the most abundant leukocytes in human blood and play a primary role in resistance against invading microorganisms and in the acute inflammatory response. However, their role in colitis and colitis-associated colorectal cancer is still under debate. This study aims to dissect the role of neutrophils in these pathologic contexts by using a rigorous genetic approach. Neutrophil-deficient mice (Csf3r-/- mice) were used in classic models of colitis and colitis-associated colorectal cancer and the role of neutrophils was assessed by histologic, cellular, and molecular analyses coupled with adoptive cell transfer. We also performed correlative analyses using human datasets. Csf3r-/- mice showed increased susceptibility to colitis and colitis-associated colorectal cancer compared with control Csf3r+/+ mice and adoptive transfer of neutrophils in Csf3r-/- mice reverted the phenotype. In colitis, Csf3r-/- mice showed increased bacterial invasion and a reduced number of healing ulcers in the colon, indicating a compromised regenerative capacity of epithelial cells. Neutrophils were essential for γδ T-cell polarization and IL22 production. In patients with ulcerative colitis, expression of CSF3R was positively correlated with IL22 and IL23 expression. Moreover, gene signatures associated with epithelial-cell development, proliferation, and antimicrobial response were enriched in CSF3Rhigh patients. Our data support a model where neutrophils mediate protection against intestinal inflammation and colitis-associated colorectal cancer by controlling the intestinal microbiota and driving the activation of an IL22-dependent tissue repair pathway.


Colitis, Ulcerative , Colitis-Associated Neoplasms , Neutrophils , Animals , Humans , Mice , Carcinogenesis , Colitis/pathology , Colitis, Ulcerative/metabolism , Colitis-Associated Neoplasms/pathology , Disease Models, Animal , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Neutrophils/metabolism
3.
Front Immunol ; 14: 1180810, 2023.
Article En | MEDLINE | ID: mdl-37180120

Neutrophils are the most abundant circulating leukocytes in humans and the first immune cells recruited at the site of inflammation. Classically perceived as short-lived effector cells with limited plasticity and diversity, neutrophils are now recognized as highly heterogenous immune cells, which can adapt to various environmental cues. In addition to playing a central role in the host defence, neutrophils are involved in pathological contexts such as inflammatory diseases and cancer. The prevalence of neutrophils in these conditions is usually associated with detrimental inflammatory responses and poor clinical outcomes. However, a beneficial role for neutrophils is emerging in several pathological contexts, including in cancer. Here we will review the current knowledge of neutrophil biology and heterogeneity in steady state and during inflammation, with a focus on the opposing roles of neutrophils in different pathological contexts.


Neoplasms , Neutrophils , Humans , Inflammation , Leukocytes/pathology
6.
Front Cell Dev Biol ; 10: 933113, 2022.
Article En | MEDLINE | ID: mdl-35874810

Tertiary lymphoid structures (TLS) are ectopic lymphoid organs that have been observed in chronic inflammatory conditions including cancer, where they are thought to exert a positive effect on prognosis. Both immune and non-immune cells participate in the genesis of TLS by establishing complex cross-talks requiring both soluble factors and cell-to-cell contact. Several immune cell types, including T follicular helper cells (Tfh), regulatory T cells (Tregs), and myeloid cells, may accumulate in TLS, possibly promoting or inhibiting their development. In this manuscript, we propose to review the available evidence regarding specific aspects of the TLS formation in solid cancers, including 1) the role of stromal cell composition and architecture in the recruitment of specific immune subpopulations and the formation of immune cell aggregates; 2) the contribution of the myeloid compartment (macrophages and neutrophils) to the development of antibody responses and the TLS formation; 3) the immunological and metabolic mechanisms dictating recruitment, expansion and plasticity of Tregs into T follicular regulatory cells, which are potentially sensitive to immunotherapeutic strategies directed to costimulatory receptors or checkpoint molecules.

7.
Cancers (Basel) ; 14(3)2022 Jan 28.
Article En | MEDLINE | ID: mdl-35158948

Neutrophils are an important component of the tumor microenvironment, and their infiltration has been associated with a poor prognosis for most human tumors. However, neutrophils have been shown to be endowed with both protumor and antitumor activities, reflecting their heterogeneity and plasticity in cancer. A growing body of studies has demonstrated that chemokines and chemokine receptors, which are fundamental regulators of neutrophils trafficking, can affect neutrophil maturation and effector functions. Here, we review human and mouse data suggesting that targeting chemokines or chemokine receptors can modulate neutrophil activity and improve their antitumor properties and the efficiency of immunotherapy.

8.
Nat Cancer ; 2(2): 218-232, 2021 02.
Article En | MEDLINE | ID: mdl-34505065

Complement has emerged as a component of tumor promoting inflammation. We conducted a systematic assessment of the role of complement activation and effector pathways in sarcomas. C3-/-, MBL1/2-/- and C4-/- mice showed reduced susceptibility to 3-methylcholanthrene sarcomagenesis and transplanted sarcomas, whereas C1q and factor B deficiency had marginal effects. Complement 3a receptor (C3aR), but not C5aR1 and C5aR2, deficiency mirrored the phenotype of C3-/- mice. C3 and C3aR deficiency were associated with reduced accumulation and functional skewing of tumor-associated macrophages, increased T cell activation and response to anti-PD-1 therapy. Transcriptional profiling of sarcoma infiltrating macrophages and monocytes revealed the enrichment of MHC II-dependent antigen presentation pathway in C3-deficient cells. In patients, C3aR expression correlated with a macrophage population signature and C3 deficiency-associated signatures predicted better clinical outcome. These results suggest that the lectin pathway and C3a/C3aR axis are key components of complement and macrophage-mediated sarcoma promotion and immunosuppression.


Lectins , Receptors, Complement/metabolism , Sarcoma , Animals , Complement Activation/physiology , Humans , Immunosuppression Therapy , Lectins/metabolism , Mice , Monocytes/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Sarcoma/drug therapy
9.
Front Immunol ; 12: 666198, 2021.
Article En | MEDLINE | ID: mdl-34093560

Klebsiella pneumoniae is a common pathogen in human sepsis. The emergence of multidrug-resistant K. pneumoniae strains represents a major clinical challenge in nosocomial and community acquired infections. The long pentraxin PTX3, a key component of humoral innate immunity, is involved in resistance to selected pathogens by promoting opsonophagocytosis. We investigated the relevance of PTX3 in innate immunity against K. pneumoniae infections using Ptx3-/- mice and mouse models of severe K. pneumoniae infections. Local and systemic PTX3 expression was induced following K. pneumoniae pulmonary infection, in association with the up-regulation of TNF-α and IL-1ß. PTX3 deficiency in mice was associated with higher bacterial burden and mortality, release of pro-inflammatory cytokines as well as IL-10 in the lung and systemically. The analysis of the mechanisms responsible of PTX3-dependent control of K. pneumoniae infection revealed that PTX3 did not interact with K. pneumoniae, or promote opsonophagocytosis. The comparison of susceptibility of wild-type, Ptx3-/-, C3-/- and Ptx3-/- /C3-/- mice to the infection showed that PTX3 acted in a complement-independent manner. Lung histopathological analysis showed more severe lesions in Ptx3-/- mice with fibrinosuppurative, necrotizing and haemorrhagic bronchopneumonia, associated with increased fibrin deposition in the lung and circulating fibrinogen consumption. These findings indicate that PTX3 contributes to the control of K. pneumoniae infection by modulating inflammatory responses and tissue damage. Thus, this study emphasizes the relevance of the role of PTX3 as regulator of inflammation and orchestrator of tissue repair in innate responses to infections.


C-Reactive Protein/immunology , Klebsiella Infections/immunology , Klebsiella pneumoniae/pathogenicity , Serum Amyloid P-Component/immunology , Animals , Bacterial Load/immunology , C-Reactive Protein/deficiency , C-Reactive Protein/metabolism , Cytokines/metabolism , Fibrin/metabolism , Fibrinogen/metabolism , Immunity, Innate , Inflammation , Klebsiella Infections/metabolism , Klebsiella Infections/microbiology , Klebsiella Infections/pathology , Klebsiella pneumoniae/immunology , Lung/immunology , Lung/metabolism , Lung/microbiology , Lung/pathology , Mice , Sepsis/immunology , Sepsis/metabolism , Sepsis/microbiology , Sepsis/pathology , Serum Amyloid P-Component/deficiency , Serum Amyloid P-Component/metabolism , Stromal Cells/metabolism
10.
Cell Mol Immunol ; 18(3): 566-578, 2021 03.
Article En | MEDLINE | ID: mdl-33473192

Myeloid cells in tumor tissues constitute a dynamic immune population characterized by a non-uniform phenotype and diverse functional activities. Both tumor-associated macrophages (TAMs), which are more abundantly represented, and tumor-associated neutrophils (TANs) are known to sustain tumor cell growth and invasion, support neoangiogenesis and suppress anticancer adaptive immune responses. In recent decades, several therapeutic approaches have been implemented in preclinical cancer models to neutralize the tumor-promoting roles of both TAMs and TANs. Some of the most successful strategies have now reached the clinic and are being investigated in clinical trials. In this review, we provide an overview of the recent literature on the ever-growing complexity of the biology of TAMs and TANs and the development of the most promising approaches to target these populations therapeutically in cancer patients.


Immunotherapy/methods , Myeloid Cells/immunology , Neoplasms/therapy , Neutrophils/immunology , Tumor-Associated Macrophages/immunology , Animals , Humans , Neoplasms/immunology
11.
Immunity ; 53(6): 1215-1229.e8, 2020 12 15.
Article En | MEDLINE | ID: mdl-33220234

Inflammation can support or restrain cancer progression and the response to therapy. Here, we searched for primary regulators of cancer-inhibitory inflammation through deep profiling of inflammatory tumor microenvironments (TMEs) linked to immune-dependent control in mice. We found that early intratumoral accumulation of interferon gamma (IFN-γ)-producing natural killer (NK) cells induced a profound remodeling of the TME and unleashed cytotoxic T cell (CTL)-mediated tumor eradication. Mechanistically, tumor-derived prostaglandin E2 (PGE2) acted selectively on EP2 and EP4 receptors on NK cells, hampered the TME switch, and enabled immune evasion. Analysis of patient datasets across human cancers revealed distinct inflammatory TME phenotypes resembling those associated with cancer immune control versus escape in mice. This allowed us to generate a gene-expression signature that integrated opposing inflammatory factors and predicted patient survival and response to immune checkpoint blockade. Our findings identify features of the tumor inflammatory milieu associated with immune control of cancer and establish a strategy to predict immunotherapy outcomes.


Immune Checkpoint Inhibitors/therapeutic use , Inflammation/immunology , Neoplasms/immunology , Tumor Escape/immunology , Animals , Dinoprostone/metabolism , Humans , Immunotherapy , Inflammation/genetics , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Mice , Neoplasms/therapy , Phenotype , Prognosis , Prostaglandin-Endoperoxide Synthases/genetics , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/immunology
12.
Semin Immunol ; 48: 101409, 2020 04.
Article En | MEDLINE | ID: mdl-32958359

Neutrophils are essential soldiers of the immune response and their role have long been restricted to their activities in defence against microbial infections and during the acute phase of the inflammatory response. However, increasing number of investigations showed that neutrophils are endowed with plasticity and can participate in the orchestration of both innate and adaptive immune responses. Neutrophils have an impact on a broad range of disorders, including infections, chronic inflammations, and cancer. Neutrophils are present in the tumour microenvironment and have been reported to mediate both pro-tumour and anti-tumour responses. Neutrophils can contribute to genetic instability, tumour cell proliferation, angiogenesis and suppression of the anti-tumour immune response. In contrast, neutrophils are reported to mediate anti-tumour resistance by direct killing of tumour cells or by engaging cooperative interactions with other immune cells. Here we discuss the current understandings of neutrophils biology and functions in health and diseases, with a specific focus on their role in cancer biology and their prognostic significance in human cancer.


Inflammation/immunology , Neoplasms/immunology , Neutrophils/immunology , Adaptive Immunity , Animals , Biodiversity , Humans , Immunity, Innate , Phenotype , Tumor Microenvironment
13.
Nat Rev Cancer ; 20(9): 485-503, 2020 09.
Article En | MEDLINE | ID: mdl-32694624

Neutrophils play a key role in defence against infection and in the activation and regulation of innate and adaptive immunity. In cancer, tumour-associated neutrophils (TANs) have emerged as an important component of the tumour microenvironment. Here, they can exert dual functions. TANs can be part of tumour-promoting inflammation by driving angiogenesis, extracellular matrix remodelling, metastasis and immunosuppression. Conversely, neutrophils can also mediate antitumour responses by direct killing of tumour cells and by participating in cellular networks that mediate antitumour resistance. Neutrophil diversity and plasticity underlie the dual potential of TANs in the tumour microenvironment. Myeloid checkpoints as well as the tumour and tissue contexture shape neutrophil function in response to conventional therapies and immunotherapy. We surmise that neutrophils can provide tools to tailor current immunotherapy strategies and pave the way to myeloid cell-centred therapeutic strategies, which would be complementary to current approaches.


Neoplasms/immunology , Neoplasms/therapy , Neutrophils/immunology , Animals , Humans , Immunotherapy , Neoplastic Processes , Tumor Microenvironment/immunology
14.
J Leukoc Biol ; 108(2): 715-721, 2020 08.
Article En | MEDLINE | ID: mdl-32108374

Kirsten rat sarcoma viral oncogene homolog KRAS proto-oncogene is the most common altered gene in colorectal cancer (CRC). Determining its mutational status, which is associated with worse prognosis and resistance to anti-epidermal growth factor receptor (EGFR) inhibitors, is essential for managing patients with CRC and colon liver metastases (CLM). Emerging studies highlighted the relationship of KRAS-mutated cancers and tumor microenvironment components, mainly with T cells. The aim of this study was to analyze the relationship of CLM immune cell infiltrate with KRAS mutational status. We performed a retrospective study on paraffin-embedded CLM tissue sections from patients surgically resected at the Department of Hepatobiliary and General Surgery of Humanitas Clinical and Cancer Center. We studied the distribution of lymphocytes (CD3+ cells), macrophages (CD163+), and neutrophils (CD66b+) in CLM tumoral and peritumoral area. Percentage of positive cells was correlated with tumor macroscopic characteristic, clinical aspects, and KRAS mutation. We observed a significant increase in CD66b+ cells in the peritumoral area in patients KRAS-mutated compared to KRAS wild-type patients. Percentages of lymphocytes and macrophages did not show significant differences. Further, neutrophils were found to be significantly increased also in the bloodstream of KRAS-mutated patients, indicating increased mobilization of neutrophils and recruitment in the CLM site. In conclusion, this study reveals a new intriguing aspect of the peritumoral microenvironment, which could pave the way for new prognostic and predictive markers for patient stratification.


Biomarkers, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Adult , Aged , Biomarkers , Female , Humans , Immunohistochemistry , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Prognosis , Proto-Oncogene Mas , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
15.
Cell ; 178(2): 346-360.e24, 2019 07 11.
Article En | MEDLINE | ID: mdl-31257026

Neutrophils are a component of the tumor microenvironment and have been predominantly associated with cancer progression. Using a genetic approach complemented by adoptive transfer, we found that neutrophils are essential for resistance against primary 3-methylcholantrene-induced carcinogenesis. Neutrophils were essential for the activation of an interferon-γ-dependent pathway of immune resistance, associated with polarization of a subset of CD4- CD8- unconventional αß T cells (UTCαß). Bulk and single-cell RNA sequencing (scRNA-seq) analyses unveiled the innate-like features and diversity of UTCαß associated with neutrophil-dependent anti-sarcoma immunity. In selected human tumors, including undifferentiated pleomorphic sarcoma, CSF3R expression, a neutrophil signature and neutrophil infiltration were associated with a type 1 immune response and better clinical outcome. Thus, neutrophils driving UTCαß polarization and type 1 immunity are essential for resistance against murine sarcomas and selected human tumors.


Disease Resistance , Neoplasms/pathology , Neutrophils/immunology , Sarcoma/pathology , T-Lymphocytes/metabolism , Animals , Chromones/toxicity , Disease Resistance/immunology , Humans , Immunity, Innate , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-12/genetics , Interleukin-12/metabolism , Kaplan-Meier Estimate , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Neoplasms/mortality , Neutrophil Infiltration , Neutrophils/cytology , Neutrophils/metabolism , Receptors, Colony-Stimulating Factor/metabolism , Sarcoma/chemically induced , Sarcoma/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Tumor Microenvironment
16.
Clin Rev Allergy Immunol ; 56(3): 308-321, 2019 Jun.
Article En | MEDLINE | ID: mdl-28963611

Sexual dimorphisms account for differences in clinical manifestations or incidence of infectious or autoimmune diseases and malignancy between females and males. Females develop enhanced innate and adaptive immune responses than males and are less susceptible to many infections of bacterial, viral, parasitic, and fungal origin and malignancies but in contrast, they are more prone to develop autoimmune diseases. The higher susceptibility to infections in males is observed from birth to adulthood, suggesting that sex chromosomes and not sex hormones have a major role in sexual dimorphism in innate immunity. Sex-based regulation of immune responses ultimately contributes to age-related disease development and life expectancy. Differences between males and females have been described in the expression of pattern recognition receptors of the innate immune response and in the functional responses of phagocytes and antigen presenting cells. Different factors have been shown to account for the sex-based disparity in immune responses, including genetic factors and hormonal mediators, which contribute independently to dimorphism in the innate immune response. For instance, several genes encoding for innate immune molecules are located on the X chromosome. In addition, estrogen and/or testosterone have been reported to modulate the differentiation, maturation, lifespan, and effector functions of innate immune cells, including neutrophils, macrophages, natural killer cells, and dendritic cells. In this review, we will focus on differences between males and females in innate immunity, which represents the first line of defense against pathogens and plays a fundamental role in the activation, regulation, and orientation of the adaptive immune response.


Immunity, Innate/physiology , Sex Characteristics , Dendritic Cells/immunology , Dendritic Cells/metabolism , Estrogens/immunology , Estrogens/metabolism , Female , Genes, X-Linked/immunology , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Menopause/immunology , Neutrophils/immunology , Neutrophils/metabolism , Pregnancy , Testosterone/immunology , Testosterone/metabolism , X-Linked Combined Immunodeficiency Diseases
17.
PLoS Pathog ; 14(12): e1007469, 2018 12.
Article En | MEDLINE | ID: mdl-30532257

Shigella spp. are pathogenic bacteria that cause bacillary dysentery in humans by invading the colonic and rectal mucosa where they induce dramatic inflammation. Here, we have analyzed the role of the soluble PRR Pentraxin 3 (PTX3), a key component of the humoral arm of innate immunity. Mice that had been intranasally infected with S. flexneri were rescued from death by treatment with recombinant PTX3. In vitro PTX3 exerts the antibacterial activity against Shigella, impairing epithelial cell invasion and contributing to the bactericidal activity of serum. PTX3 is produced upon LPS-TLR4 stimulation in accordance with the lipid A structure of Shigella. In the plasma of infected patients, the level of PTX3 amount only correlates strongly with symptom severity. These results signal PTX3 as a novel player in Shigella pathogenesis and its potential role in fighting shigellosis. Finally, we suggest that the plasma level of PTX3 in shigellosis patients could act as a biomarker for infection severity.


C-Reactive Protein/immunology , Dysentery, Bacillary/immunology , Immunity, Innate/immunology , Serum Amyloid P-Component/immunology , Shigella flexneri/immunology , Animals , Humans , Mice , Mice, Inbred C57BL
18.
Nature ; 551(7678): 110-114, 2017 11 02.
Article En | MEDLINE | ID: mdl-29072292

Interleukin-1 receptor 8 (IL-1R8, also known as single immunoglobulin IL-1R-related receptor, SIGIRR, or TIR8) is a member of the IL-1 receptor (ILR) family with distinct structural and functional characteristics, acting as a negative regulator of ILR and Toll-like receptor (TLR) downstream signalling pathways and inflammation. Natural killer (NK) cells are innate lymphoid cells which mediate resistance against pathogens and contribute to the activation and orientation of adaptive immune responses. NK cells mediate resistance against haematopoietic neoplasms but are generally considered to play a minor role in solid tumour carcinogenesis. Here we report that IL-1R8 serves as a checkpoint for NK cell maturation and effector function. Its genetic blockade unleashes NK-cell-mediated resistance to hepatic carcinogenesis, haematogenous liver and lung metastasis, and cytomegalovirus infection.


Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Liver Neoplasms/immunology , Muromegalovirus/immunology , Receptors, Interleukin-1/immunology , Animals , Cell Differentiation/genetics , Female , Herpesviridae Infections/genetics , Herpesviridae Infections/immunology , Humans , Killer Cells, Natural/metabolism , Liver Neoplasms/genetics , Lymphocyte Activation/genetics , Male , Mice , Mice, Inbred C57BL , Neoplasm Metastasis/genetics , Neoplasm Metastasis/immunology , Receptors, Interleukin-1/genetics
19.
Int Rev Immunol ; 36(3): 145-153, 2017 05 04.
Article En | MEDLINE | ID: mdl-28471269

Pentraxins are soluble pattern recognition molecules implicated not only in the opsonization and removal of microorganisms but also in the clearance of modified self (i.e., dying cells). Pentraxins can be classified into short pentraxins (C-reactive protein and serum amyloid-P component) and long pentraxins with pentraxin-3 (PTX3), the prototypic one of the latter. Pentraxins are involved in various physiological or pathophysiological processes such as inflammation and autoimmunity. A breakdown in immune tolerance to pentraxins has been reported in various autoimmune diseases. In the present review, we analyzed the current knowledge concerning anti-pentraxin antibodies, with a special focus on anti-PTX3 autoantibodies.


Autoantibodies/immunology , Autoantigens/immunology , Autoimmune Diseases/immunology , Autoimmunity , C-Reactive Protein/immunology , Serum Amyloid P-Component/immunology , Autoantigens/chemistry , Autoantigens/genetics , Autoimmune Diseases/epidemiology , Autoimmune Diseases/genetics , Autoimmune Diseases/metabolism , Biomarkers , C-Reactive Protein/chemistry , C-Reactive Protein/genetics , Comorbidity , Humans , Immune Tolerance , Ligands , Multigene Family , Prevalence , Serum Amyloid P-Component/chemistry , Serum Amyloid P-Component/genetics
20.
Emerg Top Life Sci ; 1(5): 457-470, 2017 Dec 12.
Article En | MEDLINE | ID: mdl-33525797

Neutrophils represent the most abundant leukocyte population in human peripheral blood, and their role had long been considered restricted to their phagocytic and antimicrobial activities during the acute phase of inflammation. However, an increasing number of recent investigations had highlighted their possible impact in tumor initiation and development, and the nature of neutrophil contribution in cancer had become a hot topic in immunology. Over the years, neutrophils have been shown to display both pro-tumor and antitumor effects, emphasizing an unexpected cellular heterogeneity in cancer. In this review, we will focus on the several 'shades' of neutrophils in tumor initiation, growth and metastasis. In addition, we will discuss the clinical significance of tumor-associated neutrophils in humans and their potential targeting in cancer therapy.

...