Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Proc Natl Acad Sci U S A ; 117(2): 1119-1128, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31888983

ABSTRACT

Reprogramming the tumor microenvironment to increase immune-mediated responses is currently of intense interest. Patients with immune-infiltrated "hot" tumors demonstrate higher treatment response rates and improved survival. However, only the minority of tumors are hot, and a limited proportion of patients benefit from immunotherapies. Innovative approaches that make tumors hot can have immediate impact particularly if they repurpose drugs with additional cancer-unrelated benefits. The seasonal influenza vaccine is recommended for all persons over 6 mo without prohibitive contraindications, including most cancer patients. Here, we report that unadjuvanted seasonal influenza vaccination via intratumoral, but not intramuscular, injection converts "cold" tumors to hot, generates systemic CD8+ T cell-mediated antitumor immunity, and sensitizes resistant tumors to checkpoint blockade. Importantly, intratumoral vaccination also provides protection against subsequent active influenza virus lung infection. Surprisingly, a squalene-based adjuvanted vaccine maintains intratumoral regulatory B cells and fails to improve antitumor responses, even while protecting against active influenza virus lung infection. Adjuvant removal, B cell depletion, or IL-10 blockade recovers its antitumor effectiveness. Our findings propose that antipathogen vaccines may be utilized for both infection prevention and repurposing as a cancer immunotherapy.


Subject(s)
Immunotherapy/methods , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Influenza Vaccines/therapeutic use , Injections, Intralesional , Neoplasms/drug therapy , Neoplasms/immunology , Adjuvants, Immunologic/administration & dosage , Animals , B-Lymphocytes , Basic-Leucine Zipper Transcription Factors/genetics , CD8-Positive T-Lymphocytes/immunology , Humans , Immunity, Cellular , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human , Interleukin-10 , Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Repressor Proteins/genetics , Seasons , Skin , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Squalene/administration & dosage , Tumor Microenvironment/drug effects , Vaccination
2.
Cytokine ; 138: 155404, 2021 02.
Article in English | MEDLINE | ID: mdl-33360025

ABSTRACT

The new coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can trigger a hyperinflammatory state characterized by elevated cytokine levels known as hypercytokinemia or cytokine storm, observed most often in severe patients. Though COVID-19 is known to be a primarily respiratory disease, neurological complications affecting both the central and peripheral nervous systems have also been reported. This review discusses potential routes of SARS-CoV-2 neuroinvasion and pathogenesis, summarizes reported neurological sequelae of COVID-19, and examines how aberrant cytokine levels may precipitate these complications. Clarification of the pathogenic mechanisms of SARS-CoV-2 is needed to encourage prompt diagnosis and optimized care. In particular, identifying the presence of cytokine storm in patients with neurological COVID-19 manifestations will facilitate avenues for treatment. Future investigations into aberrant cytokine levels in COVID-19 patients with neurological symptoms as well as the efficacy of cytokine storm-targeting treatments will be critical in elucidating the pathogenic mechanisms and effective treatments of COVID-19.


Subject(s)
COVID-19/pathology , Cerebrovascular Disorders/pathology , Cytokine Release Syndrome/pathology , Cytokines/blood , Nervous System Diseases/pathology , COVID-19/therapy , Central Nervous System/pathology , Cerebrovascular Disorders/virology , Cytokine Release Syndrome/therapy , Humans , Nervous System Diseases/virology , Peripheral Nervous System/pathology , SARS-CoV-2
3.
Carcinogenesis ; 36 Suppl 1: S2-18, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26106139

ABSTRACT

As part of the Halifax Project, this review brings attention to the potential effects of environmental chemicals on important molecular and cellular regulators of the cancer hallmark of evading growth suppression. Specifically, we review the mechanisms by which cancer cells escape the growth-inhibitory signals of p53, retinoblastoma protein, transforming growth factor-beta, gap junctions and contact inhibition. We discuss the effects of selected environmental chemicals on these mechanisms of growth inhibition and cross-reference the effects of these chemicals in other classical cancer hallmarks.


Subject(s)
Environmental Exposure/adverse effects , Hazardous Substances/adverse effects , Neoplasms/chemically induced , Neoplasms/etiology , Animals , Humans , Signal Transduction/drug effects
4.
Carcinogenesis ; 36 Suppl 1: S254-96, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26106142

ABSTRACT

Lifestyle factors are responsible for a considerable portion of cancer incidence worldwide, but credible estimates from the World Health Organization and the International Agency for Research on Cancer (IARC) suggest that the fraction of cancers attributable to toxic environmental exposures is between 7% and 19%. To explore the hypothesis that low-dose exposures to mixtures of chemicals in the environment may be combining to contribute to environmental carcinogenesis, we reviewed 11 hallmark phenotypes of cancer, multiple priority target sites for disruption in each area and prototypical chemical disruptors for all targets, this included dose-response characterizations, evidence of low-dose effects and cross-hallmark effects for all targets and chemicals. In total, 85 examples of chemicals were reviewed for actions on key pathways/mechanisms related to carcinogenesis. Only 15% (13/85) were found to have evidence of a dose-response threshold, whereas 59% (50/85) exerted low-dose effects. No dose-response information was found for the remaining 26% (22/85). Our analysis suggests that the cumulative effects of individual (non-carcinogenic) chemicals acting on different pathways, and a variety of related systems, organs, tissues and cells could plausibly conspire to produce carcinogenic synergies. Additional basic research on carcinogenesis and research focused on low-dose effects of chemical mixtures needs to be rigorously pursued before the merits of this hypothesis can be further advanced. However, the structure of the World Health Organization International Programme on Chemical Safety 'Mode of Action' framework should be revisited as it has inherent weaknesses that are not fully aligned with our current understanding of cancer biology.


Subject(s)
Carcinogenesis/chemically induced , Carcinogens, Environmental/adverse effects , Environmental Exposure/adverse effects , Hazardous Substances/adverse effects , Neoplasms/chemically induced , Neoplasms/etiology , Animals , Humans
5.
Mol Cancer ; 14: 137, 2015 Jul 25.
Article in English | MEDLINE | ID: mdl-26204939

ABSTRACT

From the first reported role of the transcription factor RUNX2 in osteoblast and chondrocyte differentiation and migration to its involvement in promigratory/proinvasive behavior of breast, prostate, and thyroid cancer cells, osteosarcoma, or melanoma cells, RUNX2 currently emerges as a key player in metastasis. In this review, we address the interaction of RUNX2 with the PI3K/AKT signaling pathway, one of the critical axes controlling cancer growth and metastasis. AKT, either by directly phosphorylating/activating RUNX2 or phosphorylating/inactivating regulators of RUNX2 stability or activity, contributes to RUNX2 transcriptional activity. Reciprocally, the activation of the PI3K/AKT pathway by RUNX2 regulation of its different components has been described in non-transformed and transformed cells. This mutual activation in the context of cancer cells exhibiting constitutive AKT activation and high levels of RUNX2 might constitute a major driving force in tumor progression and aggressiveness.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Core Binding Factor Alpha 1 Subunit/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Mitogen-Activated Protein Kinases/metabolism , Neoplasms/genetics , Protein Binding , Signal Transduction , Transforming Growth Factor beta/metabolism , ras Proteins/metabolism
6.
Immunology ; 142(3): 442-52, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24597649

ABSTRACT

Separate ligand-receptor paradigms are commonly used for each type of interferon (IFN). However, accumulating evidence suggests that type I and type II IFNs may not be restricted to independent pathways. Using different cell types deficient in IFNAR1, IFNAR2, IFNGR1, IFNGR2 and IFN-γ, we evaluated the contribution of each element of the IFN system to the activity of type I and type II IFNs. We show that deficiency in IFNAR1 or IFNAR2 is associated with impairment of type II IFN activity. This impairment, presumably resulting from the disruption of the ligand-receptor complex, is obtained in all cell types tested. However, deficiency of IFNGR1, IFNGR2 or IFN-γ was associated with an impairment of type I IFN activity in spleen cells only, correlating with the constitutive expression of type II IFN (IFN-γ) observed on those cells. Therefore, in vitro the constitutive expression of both the receptors and the ligands of type I or type II IFN is critical for the enhancement of the IFN activity. Any IFN deficiency can totally or partially impair IFN activity, suggesting the importance of type I and type II IFN interactions. Taken together, our results suggest that type I and type II IFNs may regulate biological activities through distinct as well as common IFN receptor complexes.


Subject(s)
Interferon Type I/immunology , Interferon Type I/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Receptor, Interferon alpha-beta/metabolism , Animals , Ligands , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/immunology
7.
J Immunother Cancer ; 11(7)2023 07.
Article in English | MEDLINE | ID: mdl-37433716

ABSTRACT

BACKGROUND: Immunotherapies are becoming front-line treatments for many advanced cancers, and combinations of two or more therapies are beginning to be investigated. Based on their individual antitumor capabilities, we sought to determine whether combination oncolytic virus (OV) and radiation therapy (RT) may improve cancer outcomes. METHODS: To investigate the activity of this combination therapy, we used in vitro mouse and human cancer cell lines as well as a mouse model of skin cancer. After initial results, we further included immune checkpoint blockade, whose addition constituted a triple combination immunotherapy. RESULTS: Our findings demonstrate that OV and RT reduce tumor growth via conversion of immunologically 'cold' tumors to 'hot', via a CD8+ T cell-dependent and IL-1α-dependent mechanism that is associated with increased PD-1/PD-L1 expression, and the triple combination of OV, RT, and PD-1 checkpoint inhibition impedes tumor growth and prolongs survival. Further, we describe the response of a PD-1-refractory patient with cutaneous squamous cell carcinoma who received the triple combination of OV, RT, and immune checkpoint inhibitor (ICI), and went on to experience unexpected, prolonged control and survival. He remains off-treatment and is without evidence of progression for >44 months since study entry. CONCLUSIONS: Effective systemic antitumor immune response is rarely elicited by a single therapy. In a skin cancer mouse model, we demonstrate improved outcomes with combination OV, RT, and ICI treatment, which is associated with mechanisms involving augmented CD8+ T cell infiltration and IL-1α expression. We report tumor reduction and prolonged survival of a patient with skin cancer treated with combination OV, RT, and ICI. Overall, our data provide strong rationale for combining OV, RT, and ICI for treatment of patients with ICI-refractory skin and potentially other cancers.


Subject(s)
Carcinoma, Squamous Cell , Immune Checkpoint Inhibitors , Oncolytic Virotherapy , Skin Neoplasms , Animals , Humans , Male , Mice , Carcinoma, Squamous Cell/therapy , Disease Models, Animal , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy , Skin Neoplasms/therapy , Cell Line, Tumor , Combined Modality Therapy
8.
Clin Dev Immunol ; 2011: 349575, 2011.
Article in English | MEDLINE | ID: mdl-22190970

ABSTRACT

The discovery of the interferon-lambda (IFN-λ) family has considerably contributed to our understanding of the role of interferon not only in viral infections but also in cancer. IFN-λ proteins belong to the new type III IFN group. Type III IFN is structurally similar to type II IFN (IFN-γ) but functionally identical to type I IFN (IFN-α/ß). However, in contrast to type I or type II IFNs, the response to type III IFN is highly cell-type specific. Only epithelial-like cells and to a lesser extent some immune cells respond to IFN-λ. This particular pattern of response is controlled by the differential expression of the IFN-λ receptor, which, in contrast to IFN-α, should result in limited side effects in patients. Recently, we and other groups have shown in several animal models a potent antitumor role of IFN-λ that will open a new challenging era for the current IFN therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Interferons/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/metabolism , Drug Synergism , Humans , Immunotherapy , Interferon-alpha/metabolism , Interferon-alpha/therapeutic use , Interferons/metabolism , Neoplasms/therapy
9.
Carcinogenesis ; 31(10): 1710-7, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20656791

ABSTRACT

Resistance to transforming growth factor (TGF) ß-mediated tumor suppression in melanoma appears to be a crucial step in tumor aggressiveness since it is usually coupled with the ability of TGFß to drive the oncogenic process via autocrine and paracrine effects. In this review, we will focus mainly on the mechanisms of escape from TGFß-induced cell cycle arrest because the mechanisms of resistance to TGFß-mediated apoptosis are still essentially speculative. As expected, some of these mechanisms can directly affect the function of the main downstream effectors of TGFß, Smad2 and Smad3, resulting in compromised Smad-mediated antiproliferative activity. Other mechanisms can counteract or overcome TGFß-mediated cell cycle arrest independently of the Smads. In melanoma, some models of resistance to TGFß have been suggested and will be described. In addition, we propose additional models of resistance taking into consideration the information available on the dysregulation of fundamental cellular effectors and signaling pathways in melanoma.


Subject(s)
Melanoma/pathology , Transforming Growth Factor beta/physiology , Apoptosis , Cell Cycle , Contractile Proteins/physiology , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase Inhibitor p21/analysis , Disease Progression , Filamins , Forkhead Box Protein O1 , Forkhead Transcription Factors/physiology , Genes, myc , Humans , Intracellular Signaling Peptides and Proteins/physiology , Melanoma/prevention & control , Microfilament Proteins/physiology , PAX3 Transcription Factor , Paired Box Transcription Factors/physiology , Phosphorylation , Proto-Oncogene Proteins/physiology , Signal Transduction , Smad2 Protein/physiology , Smad3 Protein/physiology
10.
Cancer Immunol Immunother ; 59(7): 1059-71, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20217081

ABSTRACT

Hepatocellular carcinoma (HCC) occurs most commonly secondary to cirrhosis due to chronic hepatitis C or B virus (HCV/HBV) infections. Type I interferon (IFN-alpha) treatment of chronic HCV/HBV infections reduces the incidence of HCC in cirrhotic patients. However, IFN-alpha toxicity limits its tolerability and efficacy highlighting a need for better therapeutic treatments. A recently discovered type III IFN (IFN-lambda) has been shown to possess antiviral properties against HCV and HBV in vitro. In phase I clinical trials, IFN-lambda treatment did not cause significant adverse reactions. Using a gene therapy approach, we compared the antitumor properties of IFN-alpha and IFN-lambda in a transplantable hepatoma model of HCC. BALB/c mice were inoculated with syngeneic BNL hepatoma cells, or BNL cells expressing IFN-lambda (BNL.IFN-lambda cells) or IFN-alpha (BNL.IFN-alpha cells). Despite the lack of antiproliferative activity of IFNs on BNL cells, both BNL.IFN-lambda and BNL.IFN-alpha cells displayed retarded growth kinetics in vivo. Depletion of NK cells from splenocytes inhibited splenocyte-mediated cytotoxicity, demonstrating that NK cells play a role in IFN-induced antitumor responses. However, isolated NK cells did not respond directly to IFN-lambda. There was also a marked NK cell infiltration in IFN-lambda producing tumors. In addition, IFN-lambda and, to a lesser extent, IFN-alpha enhanced immunocytotoxicity of splenocytes primed with irradiated BNL cells. Splenocyte cytotoxicity against BNL cells was dependent on IL-12 and IFN-gamma, and mediated by dendritic cells. In contrast to NK cells, isolated from spleen CD11c+ and mPDCA+ dendritic cells responded directly to IFN-lambda. The antitumor activities of IFN-lambda against hepatoma, in combination with HCV and HBV antiviral activities warrant further investigation into the clinical use of IFN-lambda to prevent HCC in HCV/HBV-infected cirrhotic patients, as well as to treat liver cancer.


Subject(s)
Cell Proliferation/drug effects , Interferon Type I/pharmacology , Interferons/pharmacology , Liver Neoplasms, Experimental/prevention & control , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cytotoxicity, Immunologic/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Female , Flow Cytometry , Immunohistochemistry , Interferon Type I/genetics , Interferons/classification , Interferons/genetics , Interleukin-12/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Liver Neoplasms, Experimental/immunology , Liver Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Transplantation , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Transfection
11.
J Interferon Cytokine Res ; 39(1): 22-29, 2019 01.
Article in English | MEDLINE | ID: mdl-30020822

ABSTRACT

Interferon (IFN)-λ, a type III interferon (IFN), is a member of a new family of pleotropic cytokines that share high similarity with classical IFNs α and ß (IFN-α/ß), type I IFNs. IFN-λ acts as an antiviral agent and displays distinct biological functions, including tumor suppression. Although it activates the common Janus kinase (JAK) and signal transducer and activator of transcription (STAT) pathways, similar to IFN-α/ß, IFN-λ differentially induces the expression of IFN-stimulated genes (ISGs). Novel evidence indicates that IFN-λ acts quite differently from IFN-α/ß under both homeostasis and pathological situations. In contrast to IFN-α/ß, IFN-λ is not involved in over-stimulation of the immune response or exacerbation of inflammation. However, the emergence of unexpected characteristics of IFN-λ, in the control of inflammation and promotion of immune suppression and cancer, reveals novel challenges and offers more strategic opportunities in the context of cancer and beyond. In this article, we discuss new evidence and potential consequences associated with the biology of IFN-λ and provide a different vision for building novel therapeutic strategies in oncology.


Subject(s)
Interferons/immunology , Neoplasms/immunology , Humans , Inflammation/immunology , Inflammation/metabolism , Interferons/metabolism , Neoplasms/metabolism , Signal Transduction/immunology
12.
Cancer Res ; 66(8): 4468-77, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16618774

ABSTRACT

Recently discovered type III IFNs (IFN-lambda) exert their antiviral and immunomodulatory activities through a unique receptor complex composed of IFN-lambdaR1 and interleukin-10 receptor 2. To further study type III IFNs, we cloned and characterized mouse IFN-lambda ligand-receptor system. We showed that, similar to their human orthologues, mIFN-lambda2 and mIFN-lambda3 signal through the IFN-lambda receptor complex, activate IFN stimulated gene factor 3, and are capable of inducing antiviral protection and MHC class I antigen expression in several cell types including B16 melanoma cells. We then used the murine B16 melanoma model to investigate the potential antitumor activities of IFN-lambdas. We developed B16 cells constitutively expressing murine IFN-lambda2 (B16.IFN-lambda2 cells) and evaluated their tumorigenicity in syngeneic C57BL/6 mice. Although constitutive expression of mIFN-lambda2 in melanoma cells did not affect their proliferation in vitro, the growth of B16.IFN-lambda2 cells, when injected s.c. into mice, was either retarded or completely prevented. We found that rejection of the modified tumor cells correlated with their level of IFN-lambda2 expression. We then developed IFN-lambda-resistant B16.IFN-lambda2 cells (B16.IFN-lambda2Res cells) and showed that their tumorigenicity was also highly impaired or completely abolished similar to B16.IFN-lambda2 cells, suggesting that IFN-lambdas engage host mechanisms to inhibit melanoma growth. These in vivo experiments show the antitumor activities of IFN-lambdas and suggest their strong therapeutic potential.


Subject(s)
Interferons/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Receptors, Interferon/immunology , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Cloning, Molecular , Cricetinae , Female , Humans , Interferons/genetics , Ligands , Melanoma, Experimental/genetics , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Receptors, Interferon/genetics , Signal Transduction , Transfection
13.
Pigment Cell Melanoma Res ; 31(2): 241-252, 2018 03.
Article in English | MEDLINE | ID: mdl-29090514

ABSTRACT

Resistance to targeted therapy in cancer is often coupled with the acquisition of a pro-invasive phenotype by tumors cells and a highly permissive tumor microenvironment promoting drug resistance. Transcription factors are frequently shown as major points of convergence of multiple dysregulated receptors and signaling pathways in cancer. Several transcription factors are now incriminated as drivers of both drug resistance and invasiveness. We focused this review on critical transcription factors playing a causal role in both the resistance to BRAF V600E-targeted therapy and the pro-invasive behavior of melanoma cells. Simultaneous rewiring of pro-oncogenic signaling pathways, phenotype switching or phenotypic plasticity supporting pro-invasive/pro-metastatic behavior, actin remodeling, and bidirectional interactions between tumor microenvironment and melanoma cells represent major challenges for overcoming resistance to BRAF V600E inhibitors (BRAFi) and will be discussed. Although it represents an underdeveloped area of translational investigation, inhibition of transcription factors may open new avenues to combat resistance to BRAFi.


Subject(s)
Drug Development , Drug Resistance, Neoplasm , Melanoma/metabolism , Melanoma/pathology , Transcription Factors/metabolism , Humans , Neoplasm Invasiveness , Signal Transduction
14.
Drug Discov Today ; 21(1): 167-171, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26552337

ABSTRACT

Interferon-lambda (IFN-λ), the most recently described type III IFN, plays a crucial part by acting on specific cell types, controlling viral infections and establishing robust innate immunity against cancer. In contrast to IFN-α or IFN-γ, IFN-λ has a restricted cell response pattern, which could make this new IFN a better choice for disease targeting and reducing adverse events. Although IFN-λ is considered to have pivotal roles in cancer, viral infections and autoimmune diseases, clinical trials have only been conducted for treatment of chronic hepatitis C virus infection. In this review, we discuss the current and the potential clinical applications of IFN-λ in the context of current IFN therapy.


Subject(s)
Interferons/pharmacology , Interferons/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Autoimmune Diseases/drug therapy , Humans , Immunity, Innate/drug effects , Neoplasms/drug therapy , Virus Diseases/drug therapy
15.
Front Immunol ; 7: 598, 2016.
Article in English | MEDLINE | ID: mdl-28018361

ABSTRACT

IFN-λ is the newly established type III IFN with unique immunomodulatory functions. In contrast to the IFN-α/ß family and to some extent IFN-γ, IFN-λ is apparently acting in specific areas of the body to activate resident immune cells and induces a local immunity, instrumental in preventing particular infections and also keeping transformed cells under control. Mucosal areas of lung and gastrointestinal tracts are now under scrutiny to elucidate the immune mechanisms triggered by IFN-λ and leading to viral protection. New evidence also indicates the crucial role of IFN-λ in promoting innate immunity in solid cancer models. Based on its unique biological activities among the IFN system, new immunotherapeutic approaches are now emerging for the treatment of cancer, infection, and autoimmune diseases. In the present review, we highlight the recent advances of IFN-λ immunomodulatory functions. We also discuss the perspectives of IFN-λ as a therapeutic agent.

16.
Immunotherapy ; 8(8): 877-88, 2016 07.
Article in English | MEDLINE | ID: mdl-27381684

ABSTRACT

Interferon-lambda (IFN-λ) is a new IFN type, related to IFN-α, that is commonly used in the clinic. However, significant side effects accompanying IFN-α treatment limit enthusiasm for IFN-α. In this review, we discuss the current landscape of IFN-α use in oncology and describe the biologic characteristics of IFN-λ. IFN-λ offers unique advantages, including a more tumor cell selective targeting, lower off-target binding and an ability to generate both innate and adaptive immune responses. IFN-λ has also demonstrated therapeutic benefit in murine cancer models. IFN-λ may be used in clinic as a single agent or in combination with other immunotherapy agents, such as immune checkpoint inhibitors. Further clinical trials will be needed to fully elucidate the potential of this novel agent in oncology.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunotherapy/methods , Interleukins/therapeutic use , Neoplasms/therapy , Adaptive Immunity , Animals , Costimulatory and Inhibitory T-Cell Receptors/antagonists & inhibitors , Disease Models, Animal , Drug Therapy, Combination , Humans , Immunity, Innate , Interferons , Mice , Neoplasms/immunology
17.
Oncotarget ; 7(31): 49259-49267, 2016 Aug 02.
Article in English | MEDLINE | ID: mdl-27363032

ABSTRACT

Hepatocellular carcinoma (HCC) is the most prevalent type of liver cancer. No significant improvement has been reported with currently available systemic therapies. IFN-α has been tested in both clinic and animal models and only moderate benefits have been observed. In animal models, similar modest antitumor efficacy has also been reported for IFN-λ, a new type of IFN that acts through its own receptor complex. In the present study, the antitumor efficacy of the combination of IFN-α and IFN-λ was tested in the BNL mouse hepatoma model. This study was accomplished by using either engineered tumor cells (IFN-α/IFN-λ gene therapy) or by directly injecting tumor-bearing mice with IFN-α/IFN-λ. Both approaches demonstrated that IFN-α/IFN-λ combination therapy was more efficacious than IFN monotherapy based on either IFN-α or IFN-λ. In complement to tumor surgery, IFN-α/IFN-λ combination induced complete tumor remission. Highest antitumor efficacy has been obtained following local administration of IFN-α/IFN-λ combination at the tumor site that was associated with strong NK cells tumor infiltration. This supports the use of IFN-α/IFN-λ combination as a new cancer immunotherapy for stimulating antitumor response after cancer surgery.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Interferon-alpha/metabolism , Interferon-gamma/metabolism , Killer Cells, Natural/cytology , Liver Neoplasms/metabolism , Animals , Carcinoma, Hepatocellular/immunology , Cell Proliferation , Cytotoxicity, Immunologic/drug effects , Female , Flow Cytometry , Genetic Therapy , Immunotherapy , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/cytology , Liver Neoplasms/immunology , Mice , Mice, Inbred BALB C
18.
Oncotarget ; 7(20): 29689-707, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27102439

ABSTRACT

Receptor tyrosine kinases-based autocrine loops largely contribute to activate the MAPK and PI3K/AKT pathways in melanoma. However, the molecular mechanisms involved in generating these autocrine loops are still largely unknown. In the present study, we examine the role of the transcription factor RUNX2 in the regulation of receptor tyrosine kinase (RTK) expression in melanoma. We have demonstrated that RUNX2-deficient melanoma cells display a significant decrease in three receptor tyrosine kinases, EGFR, IGF-1R and PDGFRß. In addition, we found co-expression of RUNX2 and another RTK, AXL, in both melanoma cells and melanoma patient samples. We observed a decrease in phosphoAKT2 (S474) and phosphoAKT (T308) levels when RUNX2 knock down resulted in significant RTK down regulation. Finally, we showed a dramatic up regulation of RUNX2 expression with concomitant up-regulation of EGFR, IGF-1R and AXL in melanoma cells resistant to the BRAF V600E inhibitor PLX4720. Taken together, our results strongly suggest that RUNX2 might be a key player in RTK-based autocrine loops and a mediator of resistance to BRAF V600E inhibitors involving RTK up regulation in melanoma.


Subject(s)
Autocrine Communication/physiology , Core Binding Factor Alpha 1 Subunit/metabolism , Gene Expression Regulation, Neoplastic/physiology , Melanoma/metabolism , Receptor Protein-Tyrosine Kinases/biosynthesis , Cell Line, Tumor , Drug Resistance, Neoplasm/physiology , Humans
19.
Cell Rep ; 17(4): 957-965, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27760326

ABSTRACT

In light of increased cancer prevalence and cancer-specific deaths in patients with infections, we investigated whether infections alter anti-tumor immune responses. We report that acute influenza infection of the lung promotes distal melanoma growth in the dermis and leads to accelerated cancer-specific host death. Furthermore, we show that during influenza infection, anti-melanoma CD8+ T cells are shunted from the tumor to the infection site, where they express high levels of the inhibitory receptor programmed cell death protein 1 (PD-1). Immunotherapy to block PD-1 reverses this loss of anti-tumor CD8+ T cells from the tumor and decreases infection-induced tumor growth. Our findings show that acute non-oncogenic infection can promote cancer growth, raising concerns regarding acute viral illness sequelae. They also suggest an unexpected role for PD-1 blockade in cancer immunotherapy and provide insight into the immune response when faced with concomitant challenges.


Subject(s)
Melanoma/immunology , Melanoma/pathology , Oncogenes , Orthomyxoviridae Infections/pathology , Acute Disease , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Lung/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL