Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Mol Cancer ; 22(1): 191, 2023 11 29.
Article in English | MEDLINE | ID: mdl-38031106

ABSTRACT

Despite major improvements in immunotherapeutic strategies, the immunosuppressive tumor microenvironment remains a major obstacle for the induction of efficient antitumor responses. In this study, we show that local delivery of a bispecific Clec9A-PD-L1 targeted type I interferon (AcTaferon, AFN) overcomes this hurdle by reshaping the tumor immune landscape.Treatment with the bispecific AFN resulted in the presence of pro-immunogenic tumor-associated macrophages and neutrophils, increased motility and maturation profile of cDC1 and presence of inflammatory cDC2. Moreover, we report empowered diversity in the CD8+ T cell repertoire and induction of a shift from naive, dysfunctional CD8+ T cells towards effector, plastic cytotoxic T lymphocytes together with increased presence of NK and NKT cells as well as decreased regulatory T cell levels. These dynamic changes were associated with potent antitumor activity. Tumor clearance and immunological memory, therapeutic immunity on large established tumors and blunted tumor growth at distant sites were obtained upon co-administration of a non-curative dose of chemotherapy.Overall, this study illuminates further application of type I interferon as a safe and efficient way to reshape the suppressive tumor microenvironment and induce potent antitumor immunity; features which are of major importance in overcoming the development of metastases and tumor cell resistance to immune attack. The strategy described here has potential for application across to a broad range of cancer types.


Subject(s)
Interferon Type I , Neoplasms , Humans , CD8-Positive T-Lymphocytes , Interferon Type I/metabolism , Tumor Microenvironment , B7-H1 Antigen/metabolism , Neoplasms/metabolism , Immunotherapy , Cell Line, Tumor
2.
Nature ; 517(7532): 89-93, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25307056

ABSTRACT

Intracellular ISG15 is an interferon (IFN)-α/ß-inducible ubiquitin-like modifier which can covalently bind other proteins in a process called ISGylation; it is an effector of IFN-α/ß-dependent antiviral immunity in mice. We previously published a study describing humans with inherited ISG15 deficiency but without unusually severe viral diseases. We showed that these patients were prone to mycobacterial disease and that human ISG15 was non-redundant as an extracellular IFN-γ-inducing molecule. We show here that ISG15-deficient patients also display unanticipated cellular, immunological and clinical signs of enhanced IFN-α/ß immunity, reminiscent of the Mendelian autoinflammatory interferonopathies Aicardi-Goutières syndrome and spondyloenchondrodysplasia. We further show that an absence of intracellular ISG15 in the patients' cells prevents the accumulation of USP18, a potent negative regulator of IFN-α/ß signalling, resulting in the enhancement and amplification of IFN-α/ß responses. Human ISG15, therefore, is not only redundant for antiviral immunity, but is a key negative regulator of IFN-α/ß immunity. In humans, intracellular ISG15 is IFN-α/ß-inducible not to serve as a substrate for ISGylation-dependent antiviral immunity, but to ensure USP18-dependent regulation of IFN-α/ß and prevention of IFN-α/ß-dependent autoinflammation.


Subject(s)
Cytokines/metabolism , Inflammation/prevention & control , Interferon Type I/immunology , Intracellular Space/metabolism , Ubiquitins/metabolism , Adolescent , Alleles , Child , Cytokines/deficiency , Cytokines/genetics , Endopeptidases/chemistry , Endopeptidases/metabolism , Female , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation/immunology , Interferon Type I/metabolism , Male , Pedigree , S-Phase Kinase-Associated Proteins/metabolism , Signal Transduction , Ubiquitin Thiolesterase , Ubiquitination , Ubiquitins/deficiency , Ubiquitins/genetics , Viruses/immunology
3.
J Autoimmun ; 97: 70-76, 2019 02.
Article in English | MEDLINE | ID: mdl-30467068

ABSTRACT

Type I Interferon (IFN) is widely used for multiple sclerosis (MS) treatment, but its side effects are limiting and its mechanism of action still unknown. Furthermore, 30-50% of MS patients are unresponsive, and IFN can even induce relapses. Fundamental understanding of the cellular target(s) of IFN will help to optimize treatments by reducing side effects and separating beneficial from detrimental effects. To improve clinical systemic IFN usage, we are developing AcTaferons (Activity-on-Target IFNs = AFNs), optimized IFN-based immunocytokines that allow cell-specific targeting. In experimental autoimmune encephalitis (EAE) in mice, high dose WT mIFNα could delay disease, but caused mortality and severe hematological deficits. In contrast, AFN targeted to dendritic cells (DC, via Clec9A) protected without mortality or hematological consequences. Conversely, CD8-targeted AFN did not protect and exacerbated weight loss, indicating the presence of both protective and unfavorable IFN effects in EAE. Comparing Clec9A-, XCR1-and SiglecH-targeting, we found that targeting AFN to plasmacytoid (p) and conventional (c) DC is superior and non-toxic compared to WT mIFN. DC-targeted AFN increased pDC numbers and their tolerogenic potential, evidenced by increased TGFß and IDO synthesis and regulatory T cell induction. In addition, both regulatory T and B cells produced significantly more immunosuppressive TGFß and IL-10. In conclusion, specific DC-targeting of IFN activity induces a robust in vivo tolerization, efficiently protecting against EAE, without noticeable side effects. Thus, dissecting positive and negative IFN effects via cell-specific targeting may result in better and safer MS therapy and response rates.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Immune Tolerance , Interferons/metabolism , Animals , B7-H1 Antigen/metabolism , Biomarkers , CTLA-4 Antigen/metabolism , Disease Models, Animal , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/pathology , Male , Mice , Models, Biological
4.
Bull Math Biol ; 80(7): 1900-1936, 2018 07.
Article in English | MEDLINE | ID: mdl-29721746

ABSTRACT

Sensing and reciprocating cellular systems (SARs) are important for the operation of many biological systems. Production in interferon (IFN) SARs is achieved through activation of the Jak-Stat pathway, and downstream upregulation of IFN regulatory factor (IRF)-7 and IFN transcription, but the role that high- and low-affinity IFNs play in this process remains unclear. We present a comparative between a minimal spatio-temporal partial differential equation model and a novel spatio-structural-temporal (SST) model for the consideration of receptor, binding, and metabolic aspects of SAR behaviour. Using the SST framework, we simulate single- and multi-cluster paradigms of IFN communication. Simulations reveal a cyclic process between the binding of IFN to the receptor, and the consequent increase in metabolism, decreasing the propensity for binding due to the internal feedback mechanism. One observes the effect of heterogeneity between cellular clusters, allowing them to individualise and increase local production, and within clusters, where we observe 'subpopular quiescence'; a process whereby intra-cluster subpopulations reduce their binding and metabolism such that other such subpopulations may augment their production. Finally, we observe the ability for low-affinity IFN to communicate a long range signal, where high affinity cannot, and the breakdown of this relationship through the introduction of cell motility. Biological systems may utilise cell motility where environments are unrestrictive and may use fixed system, with low-affinity communication, where a localised response is desirable.


Subject(s)
Models, Biological , Signal Transduction/physiology , Animals , Cell Communication , Computer Simulation , Humans , Interferons/metabolism , Ligands , Mathematical Concepts , Metabolic Networks and Pathways , Receptors, Interferon/metabolism , Spatio-Temporal Analysis
5.
Biochem J ; 446(3): 509-16, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22731491

ABSTRACT

Type I IFNs (interferons) are pathogen-induced immunoregulatory cytokines that exert anti-viral and anti-proliferative activities through binding to a common cell-surface receptor. Among the 17 human IFN subtypes, IFNß binds the IFNAR (IFNα receptor) 1/IFNAR2 receptor chains with particularly high affinity and is especially potent in select bioactivities (e.g. anti-proliferative and pro-apoptotic) when compared with IFNα2. However, no molecular basis has been ascribed to this differential action, since the two ligands are equipotent in immediate early signalling events. In the present study we report that IFNß induces Stat (signal transducer and activator of transcription) phosphorylation and transcriptional activation of ISGs (interferon-stimulated genes), including two genes with pro-apoptotic functions, for a considerably longer time frame than does IFNα2. We show that the diversification of α2/ß responses progressively builds up at the receptor level as a result of accumulating USP18 (ubiquitin specific protease 18), itself an ISG, which exerts its negative feedback action by taking advantage of the weakness of IFNα2 binding to the receptor. This represents a novel type of signalling regulation that diversifies the biological potential of IFNs α and ß.


Subject(s)
Endopeptidases/metabolism , Interferon-alpha/metabolism , Interferon-beta/metabolism , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , Binding Sites , Cell Proliferation , Humans , Interferon-alpha/genetics , Interferon-beta/genetics , Phosphorylation , Transcriptional Activation , Ubiquitin Thiolesterase
6.
J Exp Med ; 219(6)2022 06 06.
Article in English | MEDLINE | ID: mdl-35442418

ABSTRACT

Globally, autosomal recessive IFNAR1 deficiency is a rare inborn error of immunity underlying susceptibility to live attenuated vaccine and wild-type viruses. We report seven children from five unrelated kindreds of western Polynesian ancestry who suffered from severe viral diseases. All the patients are homozygous for the same nonsense IFNAR1 variant (p.Glu386*). This allele encodes a truncated protein that is absent from the cell surface and is loss-of-function. The fibroblasts of the patients do not respond to type I IFNs (IFN-α2, IFN-ω, or IFN-ß). Remarkably, this IFNAR1 variant has a minor allele frequency >1% in Samoa and is also observed in the Cook, Society, Marquesas, and Austral islands, as well as Fiji, whereas it is extremely rare or absent in the other populations tested, including those of the Pacific region. Inherited IFNAR1 deficiency should be considered in individuals of Polynesian ancestry with severe viral illnesses.


Subject(s)
Receptor, Interferon alpha-beta , Virus Diseases , Alleles , Child , Homozygote , Humans , Polynesia
7.
J Immunother Cancer ; 9(11)2021 11.
Article in English | MEDLINE | ID: mdl-34772757

ABSTRACT

BACKGROUND: Clinical success of therapeutic cancer vaccines depends on the ability to mount strong and durable antitumor T cell responses. To achieve this, potent cellular adjuvants are highly needed. Interleukin-1ß (IL-1ß) acts on CD8+ T cells and promotes their expansion and effector differentiation, but toxicity and undesired tumor-promoting side effects hamper efficient clinical application of this cytokine. METHODS: This 'cytokine problem' can be solved by use of AcTakines (Activity-on-Target cytokines), which represent fusions between low-activity cytokine mutants and cell type-specific single-domain antibodies. AcTakines deliver cytokine activity to a priori selected cell types and as such evade toxicity and unwanted off-target side effects. Here, we employ subcutaneous melanoma and lung carcinoma models to evaluate the antitumor effects of AcTakines. RESULTS: In this work, we use an IL-1ß-based AcTakine to drive proliferation and effector functionality of antitumor CD8+ T cells without inducing measurable toxicity. AcTakine treatment enhances diversity of the T cell receptor repertoire and empowers adoptive T cell transfer. Combination treatment with a neovasculature-targeted tumor necrosis factor (TNF) AcTakine mediates full tumor eradication and establishes immunological memory that protects against secondary tumor challenge. Interferon-γ was found to empower this AcTakine synergy by sensitizing the tumor microenvironment to TNF. CONCLUSIONS: Our data illustrate that anticancer cellular immunity can be safely promoted with an IL-1ß-based AcTakine, which synergizes with other immunotherapies for efficient tumor destruction.


Subject(s)
Immunotherapy/methods , Interleukin-1/metabolism , Neoplasms/immunology , Tumor Necrosis Factor-alpha/metabolism , Animals , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Disease Models, Animal , Humans , Mice
8.
Sci Rep ; 11(1): 21575, 2021 11 03.
Article in English | MEDLINE | ID: mdl-34732771

ABSTRACT

Type I Interferon (IFN) was the very first drug approved for the treatment of Multiple Sclerosis (MS), and is still frequently used as a first line therapy. However, systemic IFN also causes considerable side effects, affecting therapy adherence and dose escalation. In addition, the mechanism of action of IFN in MS is multifactorial and still not completely understood. Using AcTaferons (Activity-on-Target IFNs, AFNs), optimized IFN-based immunocytokines that allow cell-specific targeting, we have previously demonstrated that specific targeting of IFN activity to dendritic cells (DCs) can protect against experimental autoimmune encephalitis (EAE), inducing in vivo tolerogenic protective effects, evidenced by increased indoleamine-2,3-dioxygenase (IDO) and transforming growth factor ß (TGFß) release by plasmacytoid (p) DCs and improved immunosuppressive capacity of regulatory T and B cells. We here report that targeting type I IFN activity specifically towards B cells also provides strong protection against EAE, and that targeting pDCs using SiglecH-AFN can significantly add to this protective effect. The superior protection achieved by simultaneous targeting of both B lymphocytes and pDCs correlated with improved IL-10 responses in B cells and conventional cDCs, and with a previously unseen very robust IDO response in several cells, including all B and T lymphocytes, cDC1 and cDC2.


Subject(s)
B-Lymphocytes/metabolism , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/therapy , Interferons/metabolism , Animals , Antibodies/chemistry , Biotechnology , Disease Progression , Immunosuppressive Agents , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon Type I/metabolism , Lymphocyte Count , Lymphocytes/cytology , Male , Mice , Mice, Inbred C57BL , Peptides/chemistry , Signal Transduction , T-Lymphocytes/immunology , Transforming Growth Factor beta/metabolism
9.
NPJ Vaccines ; 5(1): 64, 2020.
Article in English | MEDLINE | ID: mdl-32714571

ABSTRACT

Annual administration and reformulation of influenza vaccines is required for protection against seasonal infections. However, the induction of strong and long-lasting T cells is critical to reach broad and potentially lifelong antiviral immunity. The NLRP3 inflammasome and its product interleukin-1ß (IL-1ß) are pivotal mediators of cellular immune responses to influenza, yet, overactivation of these systems leads to side effects, which hamper clinical applications. Here, we present a bypass around these toxicities by targeting the activity of IL-1ß to CD8+ T cells. Using this approach, we demonstrate safe inclusion of IL-1ß as an adjuvant in vaccination strategies, leading to full protection of mice against a high influenza virus challenge dose by raising potent T cell responses. In conclusion, this paper proposes a class of IL-1ß-based vaccine adjuvants and also provides further insight in the mechanics of cellular immune responses driven by IL-1ß.

10.
EMBO Mol Med ; 12(2): e11223, 2020 02 07.
Article in English | MEDLINE | ID: mdl-31912630

ABSTRACT

Systemic toxicities have severely limited the clinical application of tumor necrosis factor (TNF) as an anticancer agent. Activity-on-Target cytokines (AcTakines) are a novel class of immunocytokines with improved therapeutic index. A TNF-based AcTakine targeted to CD13 enables selective activation of the tumor neovasculature without any detectable toxicity in vivo. Upregulation of adhesion markers supports enhanced T-cell infiltration leading to control or elimination of solid tumors by, respectively, CAR T cells or a combination therapy with CD8-targeted type I interferon AcTakine. Co-treatment with a CD13-targeted type II interferon AcTakine leads to very rapid destruction of the tumor neovasculature and complete regression of large, established tumors. As no tumor markers are needed, safe and efficacious elimination of a broad range of tumor types becomes feasible.


Subject(s)
Immunotherapy , Neoplasms , Tumor Necrosis Factor-alpha , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/therapy
11.
Mol Cell Biol ; 26(5): 1888-97, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16479007

ABSTRACT

Alpha and beta interferons (IFN-alpha and IFN-beta) are multifunctional cytokines that exhibit differential activities through a common receptor composed of the subunits IFNAR1 and IFNAR2. Here we combined biophysical and functional studies to explore the mechanism that allows the alpha and beta IFNs to act differentially. For this purpose, we have engineered an IFN-alpha2 triple mutant termed the HEQ mutant that mimics the biological properties of IFN-beta. Compared to wild-type (wt) IFN-alpha2, the HEQ mutant confers a 30-fold higher binding affinity towards IFNAR1, comparable to that measured for IFN-beta, resulting in a much higher stability of the ternary complex as measured on model membranes. The HEQ mutant, like IFN-beta, promotes a differentially higher antiproliferative effect than antiviral activity. Both bring on a down-regulation of the IFNAR2 receptor upon induction, confirming an increased ternary complex stability of the plasma membrane. Oligonucleotide microarray experiments showed similar gene transcription profiles induced by the HEQ mutant and IFN-beta and higher levels of gene induction or repression than those for wt IFN-alpha2. Thus, we show that the differential activities of IFN-beta are directly related to the binding affinity for IFNAR1. Conservation of the residues mutated in the HEQ mutant within IFN-alpha subtypes suggests that IFN-alpha has evolved to bind IFNAR1 weakly, apparently to sustain differential levels of biological activities compared to those induced by IFN-beta.


Subject(s)
Interferon-alpha/genetics , Interferon-alpha/pharmacology , Interferon-beta/metabolism , Interferon-beta/pharmacology , Membrane Proteins/metabolism , Receptors, Interferon/metabolism , Antiviral Agents/pharmacology , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Down-Regulation , Gene Expression Regulation , Humans , Interferon-alpha/metabolism , Membrane Proteins/drug effects , Membrane Proteins/genetics , Multiprotein Complexes , Mutation , Promoter Regions, Genetic , Receptor, Interferon alpha-beta , Receptors, Interferon/drug effects , Receptors, Interferon/genetics , Transcriptional Activation
12.
J Mol Biol ; 366(2): 525-39, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-17174979

ABSTRACT

Type I interferons (IFNs) elicit antiviral, antiproliferative and immunmodulatory responses by binding to a shared cell surface receptor comprising the transmembrane proteins ifnar1 and ifnar2. Activation of differential response patterns by IFNs has been observed, suggesting that members of the family play different roles in innate immunity. The molecular basis for differential signaling has not been identified yet. Here, we have investigated the recognition of various IFNs including several human IFNalpha species, human IFNomega and human IFNbeta as well as ovine IFNtau2 by the receptor subunits in detail. Binding to the extracellular domains of ifnar1 (ifnar1-EC) and ifnar2 (ifnar2-EC) was monitored in real time by reflectance interference and total internal reflection fluorescence spectroscopy. For all IFNs investigated, competitive 1:1 interaction not only with ifnar2-EC but also with ifnar1-EC was shown. Furthermore, ternary complex formation was studied with ifnar1-EC and ifnar2-EC tethered onto solid-supported membranes. These analyses confirmed that the signaling complexes recruited by IFNs have very similar architectures. However, differences in rate and affinity constants over several orders of magnitude were observed for both the interactions with ifnar1-EC and ifnar2-EC. These data were correlated with the potencies of ISGF3 activation, antiviral and anti-proliferative activity on 2fTGH cells. The ISGF3 formation and antiviral activity correlated very well with the binding affinity towards ifnar2. In contrast, the affinity towards ifnar1 played a key role for antiproliferative activity. A striking correlation was observed for relative binding affinities towards ifnar1 and ifnar2 with the differential antiproliferative potency. This correlation was confirmed by systematically engineering IFNalpha2 mutants with very high differential antiproliferative potency.


Subject(s)
Interferon Type I/metabolism , Lipid Bilayers/metabolism , Receptors, Interferon/metabolism , Signal Transduction , Humans , Interferon Type I/chemistry , Interferon Type I/genetics , Interferon alpha-2 , Interferon-alpha/chemistry , Interferon-alpha/genetics , Ligands , Lipid Bilayers/chemistry , Models, Biological , Mutation , Protein Binding , Protein Structure, Tertiary , Receptors, Interferon/chemistry , Recombinant Proteins , Structure-Activity Relationship
13.
Biochem J ; 407(1): 141-51, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17627610

ABSTRACT

Type I IFNs (interferons) (IFNalpha/beta) form a family of related cytokines that control a variety of cellular functions through binding to a receptor composed of IFNAR (IFNalpha receptor subunit) 1 and 2. Among type I IFNs, the alpha2 and beta subtypes exhibit a large difference in their binding affinities to IFNAR1, and it was suggested that high concentrations of IFNAR1 may compensate for its low intrinsic binding affinity for IFNalpha2. We tested whether receptor-proximal signalling events are sensitive to IFNAR1 surface concentration by investigating the relationship between relative IFNAR1/IFNAR2 surface levels and IFNalpha2 and IFNbeta signalling potencies in several cell lines. For this, we monitored the activation profile of JAK (Janus kinase)/STAT (signal transducer and activator of transcription) proteins, measured basal and ligand-induced surface decay of each receptor subunit and tested the effect of variable IFNAR1 levels on IFNalpha2 signalling potency. Our data show that the cell-surface IFNAR1 level is indeed a limiting factor for assembly of the functional complex, but an increased concentration of it does not translate into an IFNalpha/beta differential JAK/STAT signalling nor does it change the dynamics of the engaged receptor. Importantly, however, our data highlight a differential effect upon routing of IFNAR2. Following binding of IFNalpha2, IFNAR2 is internalized, but, instead of being routed towards degradation as it is when complexed to IFNbeta, it recycles back to the cell surface. These observations suggest strongly that the stability and the intracellular lifetime of the ternary complex account for the differential control of IFNAR2. Moreover, the present study opens up the attractive possibility that endosomal-initiated signalling may contribute to IFNalpha/beta differential bioactivities.


Subject(s)
Down-Regulation , Interferon-alpha/pharmacology , Interferon-beta/pharmacology , Janus Kinases/metabolism , Receptor, Interferon alpha-beta/metabolism , STAT Transcription Factors/metabolism , Cells, Cultured , HeLa Cells , Humans , Jurkat Cells , Ligands , Receptor, Interferon alpha-beta/genetics , Signal Transduction , Time Factors , Transfection
14.
Oncoimmunology ; 7(3): e1398876, 2018.
Article in English | MEDLINE | ID: mdl-29399401

ABSTRACT

Despite approval for the treatment of various malignancies, clinical application of cytokines such as type I interferon (IFN) is severely impeded by their systemic toxicity. AcTakines (Activity-on-Target cytokines) are optimized immunocytokines that, when injected in mice, only reveal their activity upon cell-specific impact. We here show that type I IFN-derived AcTaferon targeted to the tumor displays strong antitumor activity without any associated toxicity, in contrast with wild type IFN. Treatment with CD20-targeted AcTaferon of CD20+ lymphoma tumors or melanoma tumors engineered to be CD20+, drastically reduced tumor growth. This antitumor effect was completely lost in IFNAR- or Batf3-deficient mice, and depended on IFN signaling in conventional dendritic cells. Also the presence of, but not the IFN signaling in, CD8+ T lymphocytes was critical for proficient antitumor effects. When combined with immunogenic chemotherapy, low-dose TNF, or immune checkpoint blockade strategies such as anti-PDL1, anti-CTLA4 or anti-LAG3, complete tumor regressions and subsequent immunity (memory) were observed, still without any concomitant morbidity, again in sharp contrast with wild type IFN. Interestingly, the combination therapy of tumor-targeted AcTaferon with checkpoint inhibiting antibodies indicated its ability to convert nonresponding tumors into responders. Collectively, our findings demonstrate that AcTaferon targeted to tumor-specific surface markers may provide a safe and generic addition to cancer (immuno)therapies.

15.
Cancer Res ; 78(2): 463-474, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29187401

ABSTRACT

An ideal generic cancer immunotherapy should mobilize the immune system to destroy tumor cells without harming healthy cells and remain active in case of recurrence. Furthermore, it should preferably not rely on tumor-specific surface markers, as these are only available in a limited set of malignancies. Despite approval for treatment of various cancers, clinical application of cytokines is still impeded by their multiple toxic side effects. Type I IFN has a long history in the treatment of cancer, but its multifaceted activity pattern and complex side effects prevent its clinical use. Here we develop AcTakines (Activity-on-Target cytokines), optimized (mutated) immunocytokines that are up to 1,000-fold more potent on target cells, allowing specific signaling in selected cell types only. Type I IFN-derived AcTaferon (AFN)-targeting Clec9A+ dendritic cells (DC) displayed strong antitumor activity in murine melanoma, breast carcinoma, and lymphoma models and against human lymphoma in humanized mice without any detectable toxic side effects. Combined with immune checkpoint blockade, chemotherapy, or low-dose TNF, complete tumor regression and long-lasting tumor immunity were observed, still without adverse effects. Our findings indicate that DC-targeted AFNs provide a novel class of highly efficient, safe, and broad-spectrum off-the-shelf cancer immunotherapeutics with no need for a tumor marker.Significance: Targeted type I interferon elicits powerful antitumor efficacy, similar to wild-type IFN, but without any toxic side effects. Cancer Res; 78(2); 463-74. ©2017 AACR.


Subject(s)
Cytokines/chemistry , Dendritic Cells/immunology , Immunotherapy , Interferon Type I/pharmacology , Mammary Neoplasms, Experimental/therapy , Melanoma, Experimental/therapy , Animals , Apoptosis , Cell Proliferation , Combined Modality Therapy , Cytokines/metabolism , Dendritic Cells/metabolism , Dendritic Cells/pathology , Female , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tumor Cells, Cultured
16.
Biochimie ; 89(6-7): 729-34, 2007.
Article in English | MEDLINE | ID: mdl-17367910

ABSTRACT

IL-28 and IL-29 were recently described as members of a new cytokine family that shares with type I interferon (IFN) the same Jak/Stat signalling pathway driving expression of a common set of genes. Accordingly, they have been named IFN lambda. IFNs lambda exhibit several common features with type I IFNs: antiviral activity, antiproliferative activity and in vivo antitumour activity. Importantly, however, IFNs lambda bind to a distinct membrane receptor, composed of IFNLR1 and IL10R2. This specific receptor usage suggests that this cytokine family does not merely replicate the type I IFN system and justifies its designation as type III IFN by the nomenclature committee of the International Society of Interferon and Cytokine Research.


Subject(s)
Cytokines/physiology , Interleukins/physiology , Animals , Antiviral Agents/pharmacology , Cell Line , Cytokines/metabolism , Genome, Human , Humans , Interferon-gamma/metabolism , Interferons/metabolism , Models, Biological , Models, Genetic , Receptors, Interleukin-10/metabolism , Signal Transduction
17.
J Leukoc Biol ; 79(6): 1286-94, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16624932

ABSTRACT

In human monocyte-derived dendritic cells (DC), infection with Mycobacterium tuberculosis and viruses or stimulation with Toll-like receptor type 3 and 4 agonists causes the release of type I interferon (IFN). Here, we describe that the IFN-beta released upon stimulation with lipopolysaccharide (LPS) or polyinosinic:polycytidylic acid (poly I:C) is responsible for a rapid and sustained signal transducer and activator of transcription 1 and 2 activation and expression of IFN-stimulated genes, such as the transcription factor IFN regulatory factor 7 and the chemokine CXC chemokine ligand 10. The autocrine production of IFN-beta from LPS and poly I:C-matured DC (mDC) induced a temporary saturation of the response to type I IFN and a marked decline in the level of the two IFN receptor (IFNAR) subunits. It is interesting that we found that upon clearing of the released cytokines, LPS-stimulated DC reacquired full responsiveness to IFN-beta but only partial responsiveness to IFN-alpha, and their maturation process was unaffected. Monitoring of surface and total levels of the receptor subunits showed that maximal expression of IFNAR2 resumed within 24 h of clearing, and IFNAR1 expression remained low. Thus, mDC can modulate their sensitivity to two IFN subtypes through a differential regulation of the IFNAR subunits.


Subject(s)
Dendritic Cells/drug effects , Gene Expression Regulation/drug effects , Interferon Inducers/pharmacology , Interferon-alpha/pharmacology , Interferon-beta/pharmacology , Membrane Proteins/biosynthesis , Receptors, Interferon/biosynthesis , Antibodies, Monoclonal/pharmacology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Chemokine CXCL10 , Chemokines, CXC/physiology , Down-Regulation/drug effects , Endotoxins/pharmacology , Humans , Interferon alpha-2 , Membrane Proteins/genetics , Monocytes/cytology , Poly I-C/pharmacology , Receptor, Interferon alpha-beta , Receptors, Interferon/genetics , Recombinant Proteins/pharmacology , STAT1 Transcription Factor/physiology , STAT2 Transcription Factor/physiology , Transcription, Genetic
18.
Mol Biol Cell ; 14(12): 4846-56, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12960428

ABSTRACT

Rho GTPases are key regulators of actin dynamics. We report that the Rho GTPase TCL, which is closely related to Cdc42 and TC10, localizes to the plasma membrane and the early/sorting endosomes in HeLa cells, suggesting a role in the early endocytic pathway. Receptor-dependent internalization of transferrin (Tf) is unaffected by suppression of endogenous TCL by small interfering RNA treatment. However, Tf accumulates in Rab5-positive uncoated endocytic vesicles and fails to reach the early endosome antigen-1-positive early endosomal compartments and the pericentriolar recycling endosomes. Moreover, Tf release upon TCL knockdown is significantly slower. Conversely, in the presence of dominant active TCL, internalized Tf accumulates in early endosome antigen-1-positive early/sorting endosomes and not in perinuclear recycling endosomes. Tf recycles directly from the early/sorting endosomes and it is normally released by the cells. The same phenotype is generated by replacing the C terminus of dominant active Cdc42 and TC10 with that of TCL, indicating that all three proteins share downstream effector proteins. Thus, TCL is essential for clathrin-dependent endocytosed receptors to enter the early/sorting endosomes. Furthermore, the active GTPase favors direct recycling from early/sorting endosomes without accumulating in the perinuclear recycling endosomes.


Subject(s)
Cell Membrane/enzymology , Endocytosis/physiology , Endosomes/enzymology , GTP Phosphohydrolases/metabolism , GTP Phosphohydrolases/drug effects , HeLa Cells , Humans , Immunohistochemistry , Protein Binding , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/physiology , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Subcellular Fractions , Transferrin/metabolism , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , rab5 GTP-Binding Proteins , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
19.
Methods Enzymol ; 410: 386-400, 2006.
Article in English | MEDLINE | ID: mdl-16938562

ABSTRACT

Quantitative polymerase chain reaction (PCR) is as old as PCR, but it has had to wait for the introduction of real-time PCR instruments to become widely used. These instruments allow monitoring of the PCR reaction on line; they involve the use of a fluorescent probe that allows quantification of the amplified DNA. Different fluorescent formats and different applications have been developed for quantitative PCR, but this chapter focuses on the use of the SYBR Green label for the quantification of specific cDNAs in reverse transcription mixes: RT-PCR. We propose optimal reaction conditions for the reactions to be performed on the different available instruments and discuss the important parameters for setting up experiments: specificity, efficiency, and reproducibility. We also introduce the reader to the problems of relative quantification.


Subject(s)
Reverse Transcriptase Polymerase Chain Reaction/methods , Animals , Humans , Oligonucleotide Array Sequence Analysis/methods
20.
J Mol Biol ; 350(3): 476-88, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15946680

ABSTRACT

Ligand-induced cross-linking of the type I interferon (IFN) receptor subunits ifnar1 and ifnar2 induces a pleiotrophic cellular response. Several studies have suggested differential signal activation by flexible recruitment of the accessory receptor subunit ifnar1. We have characterized the roles of the four Ig-like sub-domains (SDs) of the extracellular domain of ifnar1 (ifnar1-EC) for ligand recognition and receptor assembling. Various sub-fragments of ifnar1-EC were expressed in insect cells and purified to homogeneity. Solid phase binding assays with the ligands IFN(alpha)2 and IFN(beta) revealed that all three N-terminal SDs were required and sufficient for ligand binding, and that IFN(alpha)2 and IFN(beta) compete for this binding site. Cellular binding assays with different fragments, however, highlighted the key role of the membrane-proximal SD for the formation of an in situ IFN-receptor complex. Even substitution with the corresponding SD from homologous cytokine receptors did not restore high-affinity ligand binding. Receptor assembling analysis on supported lipid bilayers in vitro revealed that the membrane-proximal SD controls appropriate orientation of the receptor on the membrane, which is required for efficient association of ifnar1 into the ternary complex.


Subject(s)
Lipid Bilayers/chemistry , Membrane Proteins/chemistry , Receptors, Interferon/chemistry , Amino Acid Sequence , Base Sequence , Binding Sites , Binding, Competitive , Cell Line , Cell Membrane/metabolism , Cell Separation , Chromatography , Electrophoresis, Polyacrylamide Gel , Escherichia coli/metabolism , Flow Cytometry , Glycosylation , Green Fluorescent Proteins/metabolism , Humans , Interferon-alpha/metabolism , Interferon-beta/metabolism , Kinetics , Ligands , Lipid Bilayers/metabolism , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Folding , Protein Structure, Tertiary , Receptor, Interferon alpha-beta , Sequence Homology, Amino Acid , Signal Transduction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL