Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters

Publication year range
1.
Immunity ; 55(8): 1336-1339, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35947977

ABSTRACT

Fibroblasts strongly impact tumor progression, but whether they prime the pre-metastatic niche is poorly understood. In this issue of Immunity, Gong and Li et al. identify lung-specific immunosuppressive fibroblasts, which are hijacked by breast cancer cells to facilitate metastasis.


Subject(s)
Breast Neoplasms , Lung Neoplasms , Cell Line, Tumor , Female , Fertilizers , Fibroblasts/pathology , Humans , Lung/pathology , Lung Neoplasms/pathology , Melanoma , Neoplasm Metastasis/pathology , Skin Neoplasms , Soil , Tumor Microenvironment , Melanoma, Cutaneous Malignant
2.
Immunity ; 48(3): 399-416, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29562192

ABSTRACT

Owing to their tremendous diversity and plasticity, immune cells exert multifaceted functions in tumor-bearing hosts, ranging from anti-tumor to pro-tumor activities. Tumor immune landscapes differ greatly between and within cancer types. Emerging evidence suggests that genetic aberrations in cancer cells dictate the immune contexture of tumors. Here, we review the current understanding of the mechanisms whereby common drivers of tumorigenesis modulate the tumor immune milieu. We discuss these findings in the context of clinical observations and examine how cancer-cell-intrinsic properties can be exploited to maximize the benefit of immunomodulatory therapies. Understanding the relationship between cancer cell-intrinsic genetic events and the immune response may enable personalized immune intervention strategies for cancer patients.


Subject(s)
Disease Susceptibility/immunology , Immune System , Neoplasms/immunology , Animals , Biomarkers , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Immunomodulation , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction , Tumor Microenvironment/immunology
3.
Proc Natl Acad Sci U S A ; 120(52): e2311460120, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38127986

ABSTRACT

The TP53 gene is mutated in approximately 30% of all breast cancer cases. Adipocytes and preadipocytes, which constitute a substantial fraction of the stroma of normal mammary tissue and breast tumors, undergo transcriptional, metabolic, and phenotypic reprogramming during breast cancer development and play an important role in tumor progression. We report here that p53 loss in breast cancer cells facilitates the reprogramming of preadipocytes, inducing them to acquire a unique transcriptional and metabolic program that combines impaired adipocytic differentiation with augmented cytokine expression. This, in turn, promotes the establishment of an inflammatory tumor microenvironment, including increased abundance of Ly6C+ and Ly6G+ myeloid cells and elevated expression of the immune checkpoint ligand PD-L1. We also describe a potential gain-of-function effect of common p53 missense mutations on the inflammatory reprogramming of preadipocytes. Altogether, our study implicates p53 deregulation in breast cancer cells as a driver of tumor-supportive adipose tissue reprogramming, expanding the network of non-cell autonomous mechanisms whereby p53 dysfunction may promote cancer. Further elucidation of the interplay between p53 and adipocytes within the tumor microenvironment may suggest effective therapeutic targets for the treatment of breast cancer patients.


Subject(s)
Breast Neoplasms , Tumor Suppressor Protein p53 , Humans , Female , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Breast Neoplasms/pathology , Genes, p53 , Adipose Tissue/metabolism , Adipocytes/metabolism , Tumor Microenvironment/genetics
4.
Immunity ; 44(4): 722-4, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27096314

ABSTRACT

Although it is successful for some, most melanoma patients are refractory to T cell checkpoint inhibition. In this issue of Immunity, Merad and colleagues (2016) describe a dendritic-cell-based strategy to heighten the efficacy of therapeutic anti-PD-L1 and BRAF inhibitors in mouse melanoma models.


Subject(s)
B7-H1 Antigen , Dendritic Cells , Animals , Humans , Melanoma/immunology , T-Lymphocytes/immunology
5.
Nature ; 572(7770): 538-542, 2019 08.
Article in English | MEDLINE | ID: mdl-31367040

ABSTRACT

Cancer-associated systemic inflammation is strongly linked to poor disease outcome in patients with cancer1,2. For most human epithelial tumour types, high systemic neutrophil-to-lymphocyte ratios are associated with poor overall survival3, and experimental studies have demonstrated a causal relationship between neutrophils and metastasis4,5. However, the cancer-cell-intrinsic mechanisms that dictate the substantial heterogeneity in systemic neutrophilic inflammation between tumour-bearing hosts are largely unresolved. Here, using a panel of 16 distinct genetically engineered mouse models for breast cancer, we uncover a role for cancer-cell-intrinsic p53 as a key regulator of pro-metastatic neutrophils. Mechanistically, loss of p53 in cancer cells induced the secretion of WNT ligands that stimulate tumour-associated macrophages to produce IL-1ß, thus driving systemic inflammation. Pharmacological and genetic blockade of WNT secretion in p53-null cancer cells reverses macrophage production of IL-1ß and subsequent neutrophilic inflammation, resulting in reduced metastasis formation. Collectively, we demonstrate a mechanistic link between the loss of p53 in cancer cells, secretion of WNT ligands and systemic neutrophilia that potentiates metastatic progression. These insights illustrate the importance of the genetic makeup of breast tumours in dictating pro-metastatic systemic inflammation, and set the stage for personalized immune intervention strategies for patients with cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Inflammation/genetics , Inflammation/pathology , Neoplasm Metastasis/pathology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Wnt Proteins/metabolism , Animals , Breast Neoplasms/complications , Disease Models, Animal , Female , Inflammation/complications , Inflammation/immunology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Mice , Neutrophils/immunology
6.
Semin Immunol ; 57: 101546, 2021 10.
Article in English | MEDLINE | ID: mdl-34887163

ABSTRACT

Neutrophils are multifaceted innate immune cells that play a significant role in the progression of cancer by exerting both pro- and anti-tumorigenic functions. The crosstalk between cancer cells and neutrophils is complex and emerging evidence is pointing at cancer cell-intrinsic programs regulating neutrophil abundance, phenotype and function. Cancer cell-derived soluble mediators are key players in modulating the interaction with neutrophils. Here, we review how intrinsic features of cancer cells, including cancer cell genetics, epigenetics, signaling, and metabolism, manipulate neutrophil behavior and how to target these processes to impact cancer progression. A molecular understanding of cancer cell-intrinsic properties that shape the crosstalk with neutrophils will provide novel therapeutic strategies for personalized immunomodulation in cancer patients.


Subject(s)
Neoplasms , Neutrophils , Carcinogenesis , Humans , Immunomodulation , Signal Transduction , Tumor Microenvironment
7.
Br J Cancer ; 127(7): 1201-1213, 2022 10.
Article in English | MEDLINE | ID: mdl-35768550

ABSTRACT

BACKGROUND: Ductal carcinoma in situ (DCIS) is treated to prevent subsequent ipsilateral invasive breast cancer (iIBC). However, many DCIS lesions will never become invasive. To prevent overtreatment, we need to distinguish harmless from potentially hazardous DCIS. We investigated whether the immune microenvironment (IME) in DCIS correlates with transition to iIBC. METHODS: Patients were derived from a Dutch population-based cohort of 10,090 women with pure DCIS with a median follow-up time of 12 years. Density, composition and proximity to the closest DCIS cell of CD20+ B-cells, CD3+CD8+ T-cells, CD3+CD8- T-cells, CD3+FOXP3+ regulatory T-cells, CD68+ cells, and CD8+Ki67+ T-cells was assessed with multiplex immunofluorescence (mIF) with digital whole-slide analysis and compared between primary DCIS lesions of 77 women with subsequent iIBC (cases) and 64 without (controls). RESULTS: Higher stromal density of analysed immune cell subsets was significantly associated with higher grade, ER negativity, HER-2 positivity, Ki67 ≥ 14%, periductal fibrosis and comedonecrosis (P < 0.05). Density, composition and proximity to the closest DCIS cell of all analysed immune cell subsets did not differ between cases and controls. CONCLUSION: IME features analysed by mIF in 141 patients from a well-annotated cohort of pure DCIS with long-term follow-up are no predictors of subsequent iIBC, but do correlate with other factors (grade, ER, HER2 status, Ki-67) known to be associated with invasive recurrences.


Subject(s)
Breast Neoplasms , Carcinoma, Ductal, Breast , Carcinoma, Intraductal, Noninfiltrating , Biomarkers, Tumor/analysis , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Female , Forkhead Transcription Factors , Humans , Ki-67 Antigen , Tumor Microenvironment
8.
Nature ; 522(7556): 345-348, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-25822788

ABSTRACT

Metastatic disease remains the primary cause of death for patients with breast cancer. The different steps of the metastatic cascade rely on reciprocal interactions between cancer cells and their microenvironment. Within this local microenvironment and in distant organs, immune cells and their mediators are known to facilitate metastasis formation. However, the precise contribution of tumour-induced systemic inflammation to metastasis and the mechanisms regulating systemic inflammation are poorly understood. Here we show that tumours maximize their chance of metastasizing by evoking a systemic inflammatory cascade in mouse models of spontaneous breast cancer metastasis. We mechanistically demonstrate that interleukin (IL)-1ß elicits IL-17 expression from gamma delta (γδ) T cells, resulting in systemic, granulocyte colony-stimulating factor (G-CSF)-dependent expansion and polarization of neutrophils in mice bearing mammary tumours. Tumour-induced neutrophils acquire the ability to suppress cytotoxic T lymphocytes carrying the CD8 antigen, which limit the establishment of metastases. Neutralization of IL-17 or G-CSF and absence of γδ T cells prevents neutrophil accumulation and downregulates the T-cell-suppressive phenotype of neutrophils. Moreover, the absence of γδ T cells or neutrophils profoundly reduces pulmonary and lymph node metastases without influencing primary tumour progression. Our data indicate that targeting this novel cancer-cell-initiated domino effect within the immune system--the γδ T cell/IL-17/neutrophil axis--represents a new strategy to inhibit metastatic disease.


Subject(s)
Breast Neoplasms/pathology , Interleukin-17/biosynthesis , Neoplasm Metastasis/immunology , Neoplasm Metastasis/pathology , Neutrophils/metabolism , T-Lymphocyte Subsets/metabolism , Animals , Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Granulocyte Colony-Stimulating Factor/immunology , Granulocyte Colony-Stimulating Factor/metabolism , Interleukin-17/immunology , Interleukin-1beta/immunology , Lung/pathology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lymphatic Metastasis/immunology , Lymphatic Metastasis/pathology , Lymphocyte Activation , Mice , Neutrophils/cytology , Neutrophils/immunology , Phenotype , T-Lymphocyte Subsets/immunology , Tumor Microenvironment
9.
Nature ; 566(7745): 459-460, 2019 02.
Article in English | MEDLINE | ID: mdl-30809048
11.
Trends Immunol ; 36(4): 198-216, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25857662

ABSTRACT

Conventional anticancer therapies, such as chemotherapy, radiotherapy, and targeted therapy, are designed to kill cancer cells. However, the efficacy of anticancer therapies is not only determined by their direct effects on cancer cells but also by off-target effects within the host immune system. Cytotoxic treatment regimens elicit several changes in immune-related parameters including the composition, phenotype, and function of immune cells. Here we discuss the impact of innate and adaptive immune cells on the success of anticancer therapy. In this context we examine the opportunities to exploit host immune responses to boost tumor clearing, and highlight the challenges facing the treatment of advanced metastatic disease.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/immunology , Humans , Neoplasms/pathology
13.
Nat Rev Cancer ; 6(1): 24-37, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16397525

ABSTRACT

The main function of the mammalian immune system is to monitor tissue homeostasis, to protect against invading or infectious pathogens and to eliminate damaged cells. Therefore, it is surprising that cancer occurs with such a high frequency in humans. Recent insights that have been gained from clinical studies and experimental mouse models of carcinogenesis expand our understanding of the complex relationship between immune cells and developing tumours. Here, we examine the paradoxical role of adaptive and innate leukocytes as crucial regulators of cancer development and highlight recent insights that have been gained by manipulating immune responses in mouse models of de novo and spontaneous tumorigenesis.


Subject(s)
Immune System/physiopathology , Immunity, Innate/physiology , Leukocytes/physiology , Neoplasms/immunology , Animals , Disease Models, Animal , Humans , Immune System/pathology , Immunity, Active/physiology , Inflammation/immunology , Mice
14.
J Pathol ; 228(3): 300-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22926799

ABSTRACT

The leucine-rich repeat-containing heterotrimeric guanine nucleotide-binding protein-coupled receptor 5 (LGR5) has been identified as a marker of cycling stem cells in several epithelial tissues, including small intestine, colon, stomach and hair follicle. To investigate whether LGR5 also marks mammary epithelial stem cells, we performed in situ lineage-tracing studies and mammary gland reconstitutions with LGR5-expressing mammary epithelial cells. Interestingly, the LGR5 progeny population in mammary epithelium switches from the luminal to the myoepithelial compartment during the first 12 days of postnatal development, likely reflecting local changes in Wnt signalling. Together, our findings point to a stage-specific contribution of LGR5-expressing cells to luminal and basal epithelial lineages during postnatal mammary gland development.


Subject(s)
Cell Lineage/physiology , Epithelial Cells/cytology , Mammary Glands, Animal/growth & development , Morphogenesis/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Biomarkers/metabolism , Cell Differentiation/physiology , Cells, Cultured , Epithelial Cells/physiology , Female , Mammary Glands, Animal/cytology , Mammary Glands, Animal/physiology , Mice , Mice, Knockout , Models, Animal , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Signal Transduction/physiology , Stem Cells/cytology , Stem Cells/physiology , Wnt Proteins/physiology
15.
Cancer Cell ; 7(5): 411-23, 2005 May.
Article in English | MEDLINE | ID: mdl-15894262

ABSTRACT

Chronic inflammation predisposes tissue to cancer development; however, regulatory mechanisms underlying recruitment of innate leukocytes toward developing neoplasms are obscure. We report that genetic elimination of mature T and B lymphocytes in a transgenic mouse model of inflammation-associated de novo epithelial carcinogenesis, e.g., K14-HPV16 mice, limits neoplastic progression to development of epithelial hyperplasias that fail to recruit innate immune cells. Adoptive transfer of B lymphocytes or serum from HPV16 mice into T and B cell-deficient/HPV16 mice restores innate immune cell infiltration into premalignant tissue and reinstates necessary parameters for full malignancy, e.g., chronic inflammation, angiogenic vasculature, hyperproliferative epidermis. These findings support a model in which B lymphocytes are required for establishing chronic inflammatory states that promote de novo carcinogenesis.


Subject(s)
B-Lymphocytes/immunology , Inflammation/complications , Neoplasms, Glandular and Epithelial/etiology , Adoptive Transfer , Animals , B-Lymphocytes/cytology , B-Lymphocytes/transplantation , Blood Component Transfusion , CD4 Antigens/genetics , CD8 Antigens/genetics , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Cell Movement/immunology , Cell Proliferation , Chronic Disease , Disease Models, Animal , Gelatinases/metabolism , Granulocytes/cytology , Granulocytes/immunology , Homeodomain Proteins/genetics , Immunoglobulins/immunology , Immunoglobulins/metabolism , Inflammation/immunology , Keratinocytes/cytology , Mast Cells/cytology , Mast Cells/immunology , Mice , Mice, Knockout , Mice, Transgenic , Neoplasms, Glandular and Epithelial/immunology , Neoplasms, Glandular and Epithelial/pathology , Papillomaviridae/genetics , Skin/cytology , Skin/immunology , Vascular Endothelial Growth Factor A/metabolism
16.
Cancer Cell ; 41(3): 374-403, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36917948

ABSTRACT

Cancers represent complex ecosystems comprising tumor cells and a multitude of non-cancerous cells, embedded in an altered extracellular matrix. The tumor microenvironment (TME) includes diverse immune cell types, cancer-associated fibroblasts, endothelial cells, pericytes, and various additional tissue-resident cell types. These host cells were once considered bystanders of tumorigenesis but are now known to play critical roles in the pathogenesis of cancer. The cellular composition and functional state of the TME can differ extensively depending on the organ in which the tumor arises, the intrinsic features of cancer cells, the tumor stage, and patient characteristics. Here, we review the importance of the TME in each stage of cancer progression, from tumor initiation, progression, invasion, and intravasation to metastatic dissemination and outgrowth. Understanding the complex interplay between tumor cell-intrinsic, cell-extrinsic, and systemic mediators of disease progression is critical for the rational development of effective anti-cancer treatments.


Subject(s)
Endothelial Cells , Neoplasms , Humans , Ecosystem , Carcinogenesis , Cell Transformation, Neoplastic , Tumor Microenvironment
17.
Nat Rev Cancer ; 23(4): 193-215, 2023 04.
Article in English | MEDLINE | ID: mdl-36717668

ABSTRACT

Tumours display an astonishing variation in the spatial distribution, composition and activation state of immune cells, which impacts their progression and response to immunotherapy. Shedding light on the mechanisms that govern the diversity and function of immune cells in the tumour microenvironment will pave the way for the development of more tailored immunomodulatory strategies for the benefit of patients with cancer. Cancer cells, by virtue of their paracrine and juxtacrine communication mechanisms, are key contributors to intertumour heterogeneity in immune contextures. In this Review, we discuss how cancer cell-intrinsic features, including (epi)genetic aberrations, signalling pathway deregulation and altered metabolism, play a key role in orchestrating the composition and functional state of the immune landscape, and influence the therapeutic benefit of immunomodulatory strategies. Moreover, we highlight how targeting cancer cell-intrinsic parameters or their downstream immunoregulatory pathways is a viable strategy to manipulate the tumour immune milieu in favour of antitumour immunity.


Subject(s)
Neoplasms , Humans , Neoplasms/metabolism , Immunity , Immunotherapy , Tumor Microenvironment
18.
Oncoimmunology ; 12(1): 2201147, 2023.
Article in English | MEDLINE | ID: mdl-37089449

ABSTRACT

The clinical successes of immune checkpoint blockade (ICB) in advanced cancer patients have recently spurred the clinical implementation of ICB in the neoadjuvant and perioperative setting. However, how neoadjuvant ICB therapy affects the systemic immune landscape and metastatic spread remains to be established. Tumors promote both local and systemic expansion of regulatory T cells (Tregs), which are key orchestrators of tumor-induced immunosuppression, contributing to immune evasion, tumor progression and metastasis. Tregs express inhibitory immune checkpoint molecules and thus may be unintended targets for ICB therapy counteracting its efficacy. Using ICB-refractory models of spontaneous primary and metastatic breast cancer that recapitulate the poor ICB response of breast cancer patients, we observed that combined anti-PD-1 and anti-CTLA-4 therapy inadvertently promotes proliferation and activation of Tregs in the tumor, tumor-draining lymph node and circulation. Also in breast cancer patients, Treg levels were elevated upon ICB. Depletion of Tregs during neoadjuvant ICB in tumor-bearing mice not only reshaped the intratumoral immune landscape into a state favorable for ICB response but also induced profound and persistent alterations in systemic immunity, characterized by elevated CD8+ T cells and NK cells and durable T cell activation that was maintained after treatment cessation. While depletion of Tregs in combination with neoadjuvant ICB did not inhibit primary tumor growth, it prolonged metastasis-related survival driven predominantly by CD8+ T cells. This study demonstrates that neoadjuvant ICB therapy of breast cancer can be empowered by simultaneous targeting of Tregs, extending metastasis-related survival, independent of a primary tumor response.


Subject(s)
Breast Neoplasms , Lymphocyte Activation , T-Lymphocytes, Regulatory , Humans , Breast Neoplasms/immunology , Breast Neoplasms/therapy , T-Lymphocytes, Regulatory/immunology , Neoadjuvant Therapy , Immune Checkpoint Inhibitors/therapeutic use , Killer Cells, Natural/immunology , Myeloid Cells/immunology , Neoplasm Metastasis , Animals , Mice , CD8-Positive T-Lymphocytes/immunology
19.
Clin Cancer Res ; 29(18): 3729-3743, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37449970

ABSTRACT

PURPOSE: Patients with postpartum breast cancer diagnosed after cessation of breastfeeding (postweaning, PP-BCPW) have a particularly poor prognosis compared with patients diagnosed during lactation (PP-BCDL), or to pregnant (Pr-BC) and nulliparous (NP-BC) patients, regardless of standard prognostic characteristics. Animal studies point to a role of the involution process in stimulation of tumor growth in the mammary gland. However, in women, the molecular mechanisms that underlie this poor prognosis of patients with PP-BCPW remain vastly underexplored, due to of lack of adequate patient numbers and outcome data. EXPERIMENTAL DESIGN: We explored whether distinct prognostic features, common to all breast cancer molecular subtypes, exist in postpartum tumor tissue. Using detailed breastfeeding data, we delineated the postweaning period in PP-BC as a surrogate for mammary gland involution and performed whole transcriptome sequencing, immunohistochemical, and (multiplex) immunofluorescent analyses on tumor tissue of patients with PP-BCPW, PP-BCDL, Pr-BC, and NP-BC. RESULTS: We found that patients with PP-BCPW having a low expression level of an immunoglobulin gene signature, but high infiltration of plasma B cells, have an increased risk for metastasis and death. Although PP-BCPW tumor tissue was also characterized by an increase in CD8+ cytotoxic T cells and reduced distance among these cell types, these parameters were not associated with differential clinical outcomes among groups. CONCLUSIONS: These data point to the importance of plasma B cells in the postweaning mammary tumor microenvironment regarding the poor prognosis of PP-BCPW patients. Future prospective and in-depth research needs to further explore the role of B-cell immunobiology in this specific group of young patients with breast cancer.


Subject(s)
Breast Neoplasms , Postpartum Period , Pregnancy , Humans , Animals , Female , Lactation , Prognosis , Tumor Microenvironment/genetics
20.
Cell Metab ; 35(1): 118-133.e7, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36599297

ABSTRACT

Immunoediting sculpts immunogenicity and thwarts host anti-tumor responses in tumor cells during tumorigenesis; however, it remains unknown whether metabolic programming of tumor cells can be guided by immunosurveillance. Here, we report that T cell-mediated immunosurveillance in early-stage tumorigenesis instructs c-Myc upregulation and metabolic reprogramming in tumor cells. This previously unexplored tumor-immune interaction is controlled by non-canonical interferon gamma (IFNγ)-STAT3 signaling and supports tumor immune evasion. Our findings uncover that immunoediting instructs deregulated bioenergetic programs in tumor cells to empower them to disarm the T cell-mediated immunosurveillance by imposing metabolic tug-of-war between tumor and infiltrating T cells and forming the suppressive tumor microenvironment.


Subject(s)
Immune Evasion , Neoplasms , Humans , Neoplasms/pathology , Interferon-gamma/metabolism , T-Lymphocytes/metabolism , Carcinogenesis , Cell Transformation, Neoplastic , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL