Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
iScience ; 27(3): 109187, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38420590

RESUMO

Tissue-resident macrophages (TRMs) are abundant immune cells within pre-metastatic sites, yet their functional contributions to metastasis remain incompletely understood. Here, we show that alveolar macrophages (AMs), the main TRMs of the lung, are susceptible to downregulation of the immune stimulatory transcription factor IRF8, impairing anti-metastatic activity in models of metastatic breast cancer. G-CSF is a key tumor-associated factor (TAF) that acts upon AMs to reduce IRF8 levels and facilitate metastasis. Translational relevance of IRF8 downregulation was observed among macrophage precursors in breast cancer and a CD68hiIRF8loG-CSFhi gene signature suggests poorer prognosis in triple-negative breast cancer (TNBC), a G-CSF-expressing subtype. Our data highlight the underappreciated, pro-metastatic roles of AMs in response to G-CSF and identify the contribution of IRF8-deficient AMs to metastatic burden. AMs are an attractive target of local neoadjuvant G-CSF blockade to recover anti-metastatic activity.

2.
Immunity ; 52(2): 209-211, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32075722

RESUMO

Fever has beneficial effects on immune responses; however, its impact on T cell polarization is poorly understood. In this issue of Immunity, Wang et al. show that fever acts through a T cell-intrinsic SMAD4-dependent mechanism that selectively drives Th17 cell differentiation and pathogenicity in autoimmunity.


Assuntos
Autoimunidade , Células Th17 , Diferenciação Celular , Temperatura , Virulência
3.
Int J Hyperthermia ; 36(sup1): 22-36, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31795828

RESUMO

Purpose: The aim of this study is to investigate whether radiofrequency ablation (RFA) improves the efficacy of adoptive T cell immunotherapy in preclinical mouse cancer models.Method: Mice implanted subcutaneously (sc) with syngeneic colon adenocarcinoma or melanoma were treated with sub-curative in situ RFA (90 °C, 1 min). Trafficking of T cells to lymph nodes (LN) or tumors was quantified by homing assays and intravital microscopy (IVM) after sham procedure or RFA. Expression of trafficking molecules (CCL21 and intercellular adhesion molecule-1 [ICAM-1]) on high endothelial venules (HEV) in LN and tumor vessels was evaluated by immunofluorescence microscopy. Tumor-bearing mice were pretreated with RFA to investigate the therapeutic benefit when combined with adoptive transfer of in vitro-activated tumor-specific CD8+ T cells.Results: RFA increased trafficking of naïve CD8+ T cells to tumor-draining LN (TdLN). A corresponding increase in expression of ICAM-1 and CCL21 was detected on HEV in TdLN but not in contralateral (c)LN. IVM revealed that RFA substantially enhanced secondary firm arrest of lymphocytes selectively in HEV in TdLN. Furthermore, strong induction of ICAM-1 in tumor vessels was associated with significantly augmented trafficking of adoptively transferred in vitro-activated CD8+ T cells to tumors after RFA. Finally, preconditioning tumors with RFA augmented CD8+ T cell-mediated apoptosis of tumor targets and delayed growth of established tumors when combined with adoptive T cell transfer immunotherapy.Conclusions: These studies suggest that in addition to its role as a palliative therapeutic modality, RFA may have clinical potential as an immune-adjuvant therapy by augmenting the efficacy of adoptive T cell therapy.


Assuntos
Ablação por Radiofrequência/métodos , Linfócitos T/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Imunoterapia Adotiva , Camundongos , Camundongos Endogâmicos C57BL
4.
Handb Clin Neurol ; 156: 397-415, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30454603

RESUMO

The adaptive immune response has evolved over hundreds of millions of years to have exquisitely specific and durable mechanisms to eliminate pathogenic threats wherever they may occur in the body. Temperature has long been known to influence the response to infections, injuries, and even cancer; however, the mechanistic basis of these effects has only recently come under investigation. The picture that is emerging is that temperature can have varying impacts on adaptive immunity at multiple levels, with elevated temperatures generally promoting the activation, function, and delivery of immune cells, while reduced temperatures inhibit these processes. Here we will discuss the evidence supporting the assertion that temperature is an important modulator of adaptive immunity.


Assuntos
Imunidade Adaptativa/fisiologia , Temperatura Corporal/fisiologia , Temperatura , Animais , Humanos
5.
PLoS Genet ; 14(6): e1007368, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29879116

RESUMO

Selection pressure due to exposure to infectious pathogens endemic to Africa may explain distinct genetic variations in immune response genes. However, the impact of those genetic variations on human immunity remains understudied, especially within the context of modern lifestyles and living environments, which are drastically different from early humans in sub Saharan Africa. There are few data on population differences in constitutional immune environment, where genetic ancestry and environment are likely two primary sources of variation. In a study integrating genetic, molecular and epidemiologic data, we examined population differences in plasma levels of 14 cytokines involved in innate and adaptive immunity, including those implicated in chronic inflammation, and possible contributing factors to such differences, in 914 AA and 855 EA women. We observed significant differences in 7 cytokines, including higher plasma levels of CCL2, CCL11, IL4 and IL10 in EAs and higher levels of IL1RA and IFNα2 in AAs. Analyses of a wide range of demographic and lifestyle factors showed significant impact, with age, education level, obesity, smoking, and alcohol intake, accounting for some, but not all, observed population differences for the cytokines examined. Levels of two pro-inflammatory chemokines, CCL2 and CCL11, were strongly associated with percent of African ancestry among AAs. Through admixture mapping, the signal was pinpointed to local ancestry at 1q23, with fine-mapping analysis refined to the Duffy-null allele of rs2814778. In AA women, this variant was a major determinant of systemic levels of CCL2 (p = 1.1e-58) and CCL11 (p = 2.2e-110), accounting for 19% and 40% of the phenotypic variance, respectively. Our data reveal strong ancestral footprints in inflammatory chemokine regulation. The Duffy-null allele may indicate a loss of the buffering function for chemokine levels. The substantial immune differences by ancestry may have broad implications to health disparities between AA and EA populations.


Assuntos
Adaptação Biológica/genética , Citocinas/genética , Regulação da Expressão Gênica , Variação Genética , Seleção Genética , Imunidade Adaptativa/genética , Adulto , Alelos , Evolução Biológica , População Negra/genética , Citocinas/sangue , Sistema do Grupo Sanguíneo Duffy/genética , Meio Ambiente , Feminino , Frequência do Gene , Disparidades nos Níveis de Saúde , Voluntários Saudáveis , Humanos , Imunidade Inata/genética , Pessoa de Meia-Idade , População Branca/genética
6.
Cancer Epidemiol Biomarkers Prev ; 27(3): 321-330, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29339359

RESUMO

Background: Constitutional immunity shaped by exposure to endemic infectious diseases and parasitic worms in Sub-Saharan Africa may play a role in the etiology of breast cancer among African American (AA) women.Methods: A total of 149,514 gene variants in 433 genes across 45 immune pathways were analyzed in the AMBER consortium among 3,663 breast cancer cases and 4,687 controls. Gene-based pathway analyses were conducted using the adaptive rank truncated product statistic for overall breast cancer risk, and risk by estrogen receptor (ER) status. Unconditional logistic regression analysis was used to estimate ORs and 95% confidence intervals (CIs) for single variants.Results: The top pathways were Interleukin binding (P = 0.01), Biocarta TNFR2 (P = 0.005), and positive regulation of cytokine production (P = 0.024) for overall, ER+, and ER- cancers, respectively. The most significant gene was IL2RB (P = 0.001) for overall cancer, with rs228952 being the top variant identified (OR = 0.85; 95% CI, 0.79-0.92). Only BCL3 contained a significant variant for ER+ breast cancer. Variants in IL2RB, TLR6, IL8, PRKDC, and MAP3K1 were associated with ER- disease. The only genes showing heterogeneity between ER- and ER+ cancers were TRAF1, MAP3K1, and MAPK3 (P ≤ 0.02). We also noted genes associated with autoimmune and atopic disorders.Conclusions: Findings from this study suggest that genetic variants in immune pathways are relevant to breast cancer susceptibility among AA women, both for ER+ and ER- breast cancers.Impact: Results from this study extend our understanding of how inherited genetic variation in immune pathways is relevant to breast cancer susceptibility. Cancer Epidemiol Biomarkers Prev; 27(3); 321-30. ©2018 AACR.


Assuntos
Doenças Autoimunes/epidemiologia , Negro ou Afro-Americano/genética , Neoplasias da Mama/epidemiologia , Predisposição Genética para Doença , Mapas de Interação de Proteínas/genética , Adulto , Idoso , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Estudos de Casos e Controles , Proteína Quinase Ativada por DNA/genética , Proteína Quinase Ativada por DNA/imunologia , Proteína Quinase Ativada por DNA/metabolismo , Feminino , Humanos , Subunidade beta de Receptor de Interleucina-2/genética , Subunidade beta de Receptor de Interleucina-2/imunologia , Subunidade beta de Receptor de Interleucina-2/metabolismo , Interleucina-8/genética , Interleucina-8/imunologia , Interleucina-8/metabolismo , MAP Quinase Quinase Quinase 1/genética , MAP Quinase Quinase Quinase 1/imunologia , MAP Quinase Quinase Quinase 1/metabolismo , Pessoa de Meia-Idade , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Proteínas Nucleares/metabolismo , Polimorfismo de Nucleotídeo Único , Mapas de Interação de Proteínas/imunologia , Receptores de Estrogênio/metabolismo , Fatores de Risco , Receptor 6 Toll-Like/genética , Receptor 6 Toll-Like/imunologia , Receptor 6 Toll-Like/metabolismo
7.
Elife ; 52016 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-27929373

RESUMO

Myeloid-derived suppressor cells (MDSC) contribute to an immunosuppressive network that drives cancer escape by disabling T cell adaptive immunity. The prevailing view is that MDSC-mediated immunosuppression is restricted to tissues where MDSC co-mingle with T cells. Here we show that splenic or, unexpectedly, blood-borne MDSC execute far-reaching immune suppression by reducing expression of the L-selectin lymph node (LN) homing receptor on naïve T and B cells. MDSC-induced L-selectin loss occurs through a contact-dependent, post-transcriptional mechanism that is independent of the major L-selectin sheddase, ADAM17, but results in significant elevation of circulating L-selectin in tumor-bearing mice. Even moderate deficits in L-selectin expression disrupt T cell trafficking to distant LN. Furthermore, T cells preconditioned by MDSC have diminished responses to subsequent antigen exposure, which in conjunction with reduced trafficking, severely restricts antigen-driven expansion in widely-dispersed LN. These results establish novel mechanisms for MDSC-mediated immunosuppression that have unanticipated implications for systemic cancer immunity.


Assuntos
Imunidade Adaptativa , Tolerância Imunológica , Selectina L/biossíntese , Linfonodos/imunologia , Linfócitos/imunologia , Células Supressoras Mieloides/fisiologia , Neoplasias/fisiopatologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Regulação Neoplásica da Expressão Gênica , Linfócitos/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias/imunologia , Interferência de RNA , Transplante Heterólogo
8.
Oncoimmunology ; 5(5): e1116675, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27467928

RESUMO

Promising cancer immunotherapeutics depend on mobilization of cytotoxic T cells across tumor vascular barriers through mechanisms that are poorly understood. Recently, we discovered that the CXCR3 chemokine receptor uniquely functions as the master-regulator of cytotoxic CD8(+) T cell extravasation and tumor control despite the multiplicity of chemokines available in the tumor landscape.

9.
Nat Commun ; 7: 10684, 2016 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-26883450

RESUMO

Tumour vessels have been studied extensively as they are critical sites for drug delivery, anti-angiogenic therapies and immunotherapy. As a preclinical tool, intravital microscopy (IVM) allows for in vivo real-time direct observation of vessels at the cellular level. However, to date there are no reports of intravital high-resolution imaging of human tumours in the clinical setting. Here we report the feasibility of IVM examinations of human malignant disease with an emphasis on tumour vasculature as the major site of tumour-host interactions. Consistent with preclinical observations, we show that patient tumour vessels are disorganized, tortuous and ∼50% do not support blood flow. Human tumour vessel diameters are larger than predicted from immunohistochemistry or preclinical IVM, and thereby have lower wall shear stress, which influences delivery of drugs and cellular immunotherapies. Thus, real-time clinical imaging of living human tumours is feasible and allows for detection of characteristics within the tumour microenvironment.


Assuntos
Vasos Sanguíneos/química , Microscopia Intravital/métodos , Melanoma/química , Animais , Linhagem Celular Tumoral , Humanos , Melanoma/irrigação sanguínea , Melanoma/fisiopatologia , Camundongos , Microcirculação
10.
J Vaccines Vaccin ; 7(2)2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28944091

RESUMO

A lack of effective immune response against cancer is one of the major risk factors for developing local recurrence and distant metastases after curative resectional surgery. Prior studies revealed that systemic antitumor immunity is elicited by radiofrequency ablation (RFA) of tumor lesions, which is mainly considered a palliative procedure for unresectable tumors or for inoperable patients. Recently, we discovered an oncological benefit that depends on the adaptive arm of the antitumor immune response when RFA is performed in a neoadjuvant setting prior to surgical resection in preclinical murine models.

11.
PLoS One ; 10(11): e0143370, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26599402

RESUMO

PURPOSE: While surgical resection is a cornerstone of cancer treatment, local and distant recurrences continue to adversely affect outcome in a significant proportion of patients. Evidence that an alternative debulking strategy involving radiofrequency ablation (RFA) induces antitumor immunity prompted the current investigation of the efficacy of performing RFA prior to surgical resection (pre-resectional RFA) in a preclinical mouse model. EXPERIMENTAL DESIGN: Therapeutic efficacy and systemic immune responses were assessed following pre-resectional RFA treatment of murine CT26 colon adenocarcinoma. RESULTS: Treatment with pre-resectional RFA significantly delayed tumor growth and improved overall survival compared to sham surgery, RFA, or resection alone. Mice in the pre-resectional RFA group that achieved a complete response demonstrated durable antitumor immunity upon tumor re-challenge. Failure to achieve a therapeutic benefit in immunodeficient mice confirmed that tumor control by pre-resectional RFA depends on an intact adaptive immune response rather than changes in physical parameters that make ablated tumors more amenable to a complete surgical excision. RFA causes a marked increase in intratumoral CD8+ T lymphocyte infiltration, thus substantially enhancing the ratio of CD8+ effector T cells: FoxP3+ regulatory T cells. Importantly, pre-resectional RFA significantly increases the number of antigen-specific CD8+ T cells within the tumor microenvironment and tumor-draining lymph node but had no impact on infiltration by myeloid-derived suppressor cells, M1 macrophages or M2 macrophages at tumor sites or in peripheral lymphoid organs (i.e., spleen). Finally, pre-resectional RFA of primary tumors delayed growth of distant tumors through a mechanism that depends on systemic CD8+ T cell-mediated antitumor immunity. CONCLUSION: Improved survival and antitumor systemic immunity elicited by pre-resectional RFA support the translational potential of this neoadjuvant treatment for cancer patients with high-risk of local and systemic recurrence.


Assuntos
Imunidade Adaptativa/efeitos dos fármacos , Adjuvantes Imunológicos/farmacologia , Ablação por Cateter , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/cirurgia , Recidiva Local de Neoplasia/prevenção & controle , Animais , Antígenos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/patologia , Citocinas/metabolismo , Feminino , Linfonodos/efeitos dos fármacos , Linfonodos/patologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Modelos Biológicos , Invasividade Neoplásica , Recidiva Local de Neoplasia/patologia , Análise de Sobrevida , Microambiente Tumoral/efeitos dos fármacos
12.
Nat Rev Immunol ; 15(6): 335-49, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25976513

RESUMO

Fever is a cardinal response to infection that has been conserved in warm-blooded and cold-blooded vertebrates for more than 600 million years of evolution. The fever response is executed by integrated physiological and neuronal circuitry and confers a survival benefit during infection. In this Review, we discuss our current understanding of how the inflammatory cues delivered by the thermal element of fever stimulate innate and adaptive immune responses. We further highlight the unexpected multiplicity of roles of the pyrogenic cytokine interleukin-6 (IL-6), both during fever induction and during the mobilization of lymphocytes to the lymphoid organs that are the staging ground for immune defence. We also discuss the emerging evidence suggesting that the adrenergic signalling pathways associated with thermogenesis shape immune cell function.


Assuntos
Regulação da Temperatura Corporal/imunologia , Febre/imunologia , Imunidade Inata/imunologia , Animais , Citocinas/biossíntese , Proteínas de Choque Térmico/imunologia , Homeostase/imunologia , Humanos , Infecções/imunologia , Interleucina-6/imunologia , Células Matadoras Naturais/imunologia , Linfócitos/imunologia , Macrófagos/imunologia , Estresse Fisiológico/imunologia
13.
Semin Immunol ; 26(1): 38-47, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24602448

RESUMO

Within the tumor microenvironment, IL-6 signaling is generally considered a malevolent player, assuming a dark visage that promotes tumor progression. Chronic IL-6 signaling is linked to tumorigenesis in numerous mouse models as well as in human disease. IL-6 acts intrinsically on tumor cells through numerous downstream mediators to support cancer cell proliferation, survival, and metastatic dissemination. Moreover, IL-6 can act extrinsically on other cells within the complex tumor microenvironment to sustain a pro-tumor milieu by supporting angiogenesis and tumor evasion of immune surveillance. A lesser known role for IL-6 signaling has recently emerged in which it plays a beneficial role, presenting a fairer face that opposes tumor growth by mobilizing anti-tumor T cell immune responses to attain tumor control. Accumulating evidence establishes IL-6 as a key player in the activation, proliferation and survival of lymphocytes during active immune responses. IL-6 signaling can also resculpt the T cell immune response, shifting it from a suppressive to a responsive state that can effectively act against tumors. Finally, IL-6 plays an indispensable role in boosting T cell trafficking to lymph nodes and to tumor sites, where they have the opportunity to become activated and execute their cytotoxic effector functions, respectively. Here, we discuss the dual faces of IL-6 signaling in the tumor microenvironment; the dark face that drives malignancy, and the fairer aspect that promotes anti-tumor adaptive immunity.


Assuntos
Interleucina-6/metabolismo , Neoplasias/metabolismo , Microambiente Tumoral , Imunidade Adaptativa , Animais , Movimento Celular/genética , Movimento Celular/imunologia , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/imunologia , Transformação Celular Neoplásica/metabolismo , Microambiente Celular/genética , Microambiente Celular/imunologia , Humanos , Interleucina-6/genética , Linfonodos/imunologia , Linfonodos/metabolismo , Neoplasias/genética , Neoplasias/imunologia , Transdução de Sinais , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
14.
J Immunol ; 191(8): 4348-57, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24026079

RESUMO

Blood-borne neutrophils are excluded from entering lymph nodes across vascular portals termed high endothelial venules (HEVs) because of lack of expression of the CCR7 homeostatic chemokine receptor. Induction of sterile inflammation increases neutrophil entry into tumor-draining lymph nodes (TDLNs), which is critical for induction of antitumor adaptive immunity following treatments such as photodynamic therapy (PDT). However, the mechanisms controlling neutrophil entry into TDLNs remain unclear. Prior evidence that IL-17 promotes neutrophil emigration to sites of infection via induction of CXCL2 and CXCL1 inflammatory chemokines raised the question of whether IL-17 contributes to chemokine-dependent trafficking in TDLNs. In this article, we demonstrate rapid accumulation of IL-17-producing Th17 cells in the TDLNs following induction of sterile inflammation by PDT. We further report that nonhematopoietic expression of IL-17RA regulates neutrophil accumulation in TDLNs following induction of sterile inflammation by PDT. We show that HEVs are the major route of entry of blood-borne neutrophils into TDLNs through interactions of l-selectin with HEV-expressed peripheral lymph node addressin and by preferential interactions between CXCR2 and CXCL2 but not CXCL1. CXCL2 induction in TDLNs was mapped in a linear pathway downstream of IL-17RA-dependent induction of IL-1ß. These results define a novel IL-17-dependent mechanism promoting neutrophil delivery across HEVs in TDLNs during acute inflammatory responses.


Assuntos
Inflamação/imunologia , Interleucina-17/metabolismo , Linfonodos/imunologia , Neoplasias/imunologia , Neutrófilos/metabolismo , Animais , Movimento Celular/imunologia , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/biossíntese , Quimiocina CXCL2/metabolismo , Feminino , Interleucina-1beta/biossíntese , Selectina L/metabolismo , Linfonodos/citologia , Linfonodos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Neutrófilos/imunologia , Fotoquimioterapia , Receptores de Interleucina-17/biossíntese , Receptores de Interleucina-8B/metabolismo , Células Th17/imunologia
15.
Int J Hyperthermia ; 29(5): 464-73, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23862980

RESUMO

Cancer immunotherapy aims to generate long-lived, tumour-specific adaptive immunity to limit dysregulated tumour progression and metastasis. Tumour vasculature has emerged as a critical checkpoint controlling the efficacy of immunotherapy since it is the main access point for cytotoxic T cells to reach tumour cell targets. Therapeutic success has been particularly challenging to achieve because of the local, cytokine-rich inflammatory milieu that drives a pro-tumourigenic programme supporting the growth and survival of malignant cells. Here, we focus on recent evidence that systemic thermal therapy can switch the activities of the inflammatory cytokine, interleukin-6 (IL-6), to a predominantly anti-tumourigenic function that promotes anti-tumour immunity by mobilising T cell trafficking in the recalcitrant tumour microenvironment.


Assuntos
Hipertermia Induzida , Interleucina-6/imunologia , Neoplasias/terapia , Animais , Linfócitos T CD8-Positivos/imunologia , Humanos , Imunoterapia Adotiva , Neoplasias/imunologia , Microambiente Tumoral/imunologia
16.
Breast Cancer Res Treat ; 139(2): 477-88, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23624818

RESUMO

Immune signatures in breast tumors differ by estrogen receptor (ER) status. The purpose of this study was to assess associations between ER phenotypes and circulating levels of cytokines that co-ordinate cell-mediated [T-helper type 1 (Th1)] and humoral [T-helper type 2 (Th2)] immunity. We conducted a case-case comparison of 523 women with newly diagnosed breast cancer to evaluate associations between 27 circulating cytokines, measured using Luminex XMap technology, and breast cancer phenotypes [ER(-) vs. ER(+); triple negative breast cancer (TNBC) vs. luminal A (LumA)]. Ratios of Th1 to Th2 cytokines were also evaluated. Levels of interleukin (IL)-5, a Th-2 cytokine, were higher in ER(-) than in ER(+) tumors. The highest tertile of IL-5 was more strongly associated with ER(-) (OR = 2.33, 95 % CI 1.40-3.90) and TNBCs (OR = 2.78, 95 % CI 1.53-5.06) compared to ER(+) and LumA cancers, respectively, particularly among premenopausal women (OR = 4.17, 95 % CI 1.86-9.34, ER(-) vs. ER(+); OR = 5.60, 95 % CI 2.09-15.01, TNBC vs. LumA). Elevated Th1 cytokines were also detected in women with ER(-) and TNBCs, with women in the highest tertile of interferon α2 (OR = 2.39, 95 % CI 1.31-4.35) or tumor necrosis factor-α (OR = 2.27, 95 % CI 1.21-4.26) being twice as likely to have TNBC versus LumA cancer. When cytokine ratios were examined, women with the highest ratios of Th1 cytokines to IL-5 levels were least likely to have ER(-) or TNBCs compared to ER(+) or LumA cancers, respectively. The strongest associations were in premenopausal women, who were up to 80 % less likely to have TNBC than LumA cancers (IL-12p40/IL-5, OR = 0.19, 95 % CI 0.07-0.56). These findings indicate that immune function is associated with ER(-) and TNBC and may be most relevant among younger women, who are likely to be diagnosed with these aggressive phenotypes.


Assuntos
Neoplasias da Mama/metabolismo , Citocinas/sangue , Neoplasias de Mama Triplo Negativas/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Feminino , Humanos , Pessoa de Meia-Idade , Gradação de Tumores , Estadiamento de Neoplasias , Receptores de Estrogênio , Células Th1/imunologia , Células Th1/metabolismo , Células Th2/imunologia , Células Th2/metabolismo , Neoplasias de Mama Triplo Negativas/sangue , Neoplasias de Mama Triplo Negativas/imunologia
17.
Cancer Immunol Res ; 1(4): 210-6, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24490177

RESUMO

A major goal of cancer immunology is to stimulate the generation of long-lasting, tumor antigen-specific immune responses that recognize and destroy tumor cells. This article discusses advances in thermal medicine with the potential to improve cancer immunotherapy. Accumulating evidence indicates that survival benefits are accorded to individuals who achieve an increase in body temperature (i.e. fever) following infection. Furthermore, accumulating evidence indicates that physiological responses to hyperthermia impact the tumor microenvironment through temperature-sensitive check-points that regulate tumor vascular perfusion, lymphocyte trafficking, inflammatory cytokine expression, tumor metabolism, and innate and adaptive immune function. Nevertheless, the influence of thermal stimuli on the immune system, particularly the antitum or immune response, remains incompletely understood. In fact, temperature is still rarely considered as a critical variable in experimental immunology. We suggest that more attention should be directed to the role of temperature in the regulation of the immune response and that thermal therapy should be tested in conjunction with immunotherapy as a multi-functional adjuvant that modulates the dynamics of the tumor microenvironment.


Assuntos
Febre/imunologia , Sistema Imunitário/fisiologia , Neoplasias/imunologia , Temperatura , Animais , Hipóxia Celular , Citocinas/imunologia , Humanos , Hipertermia Induzida , Imunoterapia , Camundongos , Microambiente Tumoral
18.
J Clin Invest ; 121(10): 3846-59, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21926464

RESUMO

Immune cells are key regulators of neoplastic progression, which is often mediated through their release of cytokines. Inflammatory cytokines such as IL-6 exert tumor-promoting activities by driving growth and survival of neoplastic cells. However, whether these cytokines also have a role in recruiting mediators of adaptive anticancer immunity has not been investigated. Here, we report that homeostatic trafficking of tumor-reactive CD8+ T cells across microvascular checkpoints is limited in tumors despite the presence of inflammatory cytokines. Intravital imaging in tumor-bearing mice revealed that systemic thermal therapy (core temperature elevated to 39.5°C ± 0.5°C for 6 hours) activated an IL-6 trans-signaling program in the tumor blood vessels that modified the vasculature such that it could support enhanced trafficking of CD8+ effector/memory T cells (Tems) into tumors. A concomitant decrease in tumor infiltration by Tregs during systemic thermal therapy resulted in substantial enhancement of Tem/Treg ratios. Mechanistically, IL-6 produced by nonhematopoietic stromal cells acted cooperatively with soluble IL-6 receptor-α and thermally induced gp130 to promote E/P-selectin- and ICAM-1-dependent extravasation of cytotoxic T cells in tumors. Parallel increases in vascular adhesion were induced by IL-6/soluble IL-6 receptor-α fusion protein in mouse tumors and patient tumor explants. Finally, a causal link was established between IL-6-dependent licensing of tumor vessels for Tem trafficking and apoptosis of tumor targets. These findings suggest that the unique IL-6-rich tumor microenvironment can be exploited to create a therapeutic window to boost T cell-mediated antitumor immunity and immunotherapy.


Assuntos
Interleucina-6/metabolismo , Neoplasias/irrigação sanguínea , Neoplasias/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Apoptose , Linhagem Celular Tumoral , Movimento Celular/imunologia , Selectina E/metabolismo , Humanos , Hipertermia Induzida , Molécula 1 de Adesão Intercelular/metabolismo , Camundongos , Microvasos/imunologia , Modelos Imunológicos , Neoplasias/patologia , Neoplasias/terapia , Selectina-P/metabolismo , Transdução de Sinais , Microambiente Tumoral/imunologia
19.
Cancer Res ; 71(11): 3872-80, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21512134

RESUMO

Human and rodent solid tumors often exhibit elevated interstitial fluid pressure (IFP). This condition is recognized as a prognostic indicator for reduced responses to therapy and decreased disease-free survival rate. In the present study, we tested whether induction of a thermoregulatory-mediated increase in tissue blood flow, induced by exposure of mice to mild environmental heat stress, could influence IFP and other vascular parameters within tumors. Using several murine tumor models, we found that heating results in a sustained reduction in tumor IFP correlating with increased tumor vascular perfusion (measured by fluorescent imaging of perfused vessels, laser Doppler flowmetry, and MRI) as well as a sustained reduction in tumor hypoxia. Furthermore, when radiation therapy was administered 24 hours postheating, we observed a significant improvement in efficacy that may be a result of the sustained reduction in tumor hypoxia. These data suggest, for the first time, that environmental manipulation of normal vasomotor function is capable of achieving therapeutically beneficial changes in IFP and microvascular function in the tumor microenvironment.


Assuntos
Neoplasias do Colo/terapia , Hipertermia Induzida/métodos , Neoplasias Mamárias Experimentais/terapia , Melanoma Experimental/terapia , Animais , Temperatura Corporal/fisiologia , Hipóxia Celular/fisiologia , Neoplasias do Colo/irrigação sanguínea , Neoplasias do Colo/radioterapia , Terapia Combinada , Modelos Animais de Doenças , Líquido Extracelular/fisiologia , Feminino , Humanos , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/radioterapia , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/radioterapia , Camundongos , Camundongos Endogâmicos BALB C
20.
Ann Surg Oncol ; 18(8): 2357-63, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21301970

RESUMO

BACKGROUND: Although often proposed as a means to reduce the harmful consequences of tumor spill, water lavage has yet to be systematically evaluated in relevant in vitro and in vivo models. This study evaluates the mechanisms and utility of a single water lavage to improve the sequelae of tumor spill during laparotomy. METHODS: Murine colorectal tumor cell susceptibility to water-induced osmotic lysis was characterized in vitro. A reproducible model of tumor spill was established to recapitulate water or saline lavage during laparotomy. Analyses of tumor volumes calculated from noninvasive imaging were performed. The tumor volumes and survival of mice treated with water, normal saline, or sham laparotomy were assessed. RESULTS: Significant osmotic lysis of cultured murine colorectal cancer cells was observed after a brief exposure to water. Compared to saline or sham laparotomy, water lavage demonstrated superior clinical outcomes with a decrease in tumor burden and concomitant improvement in survival. CONCLUSIONS: The use of water lavage during oncologic surgeries to reduce the sequelae of tumor spill is justified and strongly supported by our study. Data from our study raise several concerns regarding the mechanisms and efficacy of saline lavage. Clinically, the use of water lavage during laparotomy would be anticipated to reduce peritoneal disease burden with minimal toxicity or cost.


Assuntos
Neoplasias Colorretais/patologia , Neoplasias Colorretais/prevenção & controle , Irrigação Terapêutica , Água , Animais , Sobrevivência Celular , Laparotomia , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos BALB C , Lavagem Peritoneal , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...