Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Med Chem ; 65(5): 4350-4366, 2022 03 10.
Article in English | MEDLINE | ID: mdl-35195996

ABSTRACT

Mutations in MEK1/2 have been described as a resistance mechanism to BRAF/MEK inhibitor treatment. We report the discovery of a novel ATP-competitive MEK1/2 inhibitor with efficacy in wildtype (WT) and mutant MEK12 models. Starting from a HTS hit, we obtained selective, cellularly active compounds that showed equipotent inhibition of WT MEK1/2 and a panel of MEK1/2 mutant cell lines. Using a structure-based approach, the optimization addressed the liabilities by systematic analysis of molecular matched pairs (MMPs) and ligand conformation. Addition of only three heavy atoms to early tool compound 6 removed Cyp3A4 liabilities and increased the cellular potency by 100-fold, while reducing log P by 5 units. Profiling of MAP855, compound 30, in pharmacokinetic-pharmacodynamic and efficacy studies in BRAF-mutant models showed comparable efficacy to clinical MEK1/2 inhibitors. Compound 30 is a novel highly potent and selective MEK1/2 kinase inhibitor with equipotent inhibition of WT and mutant MEK1/2, whose drug-like properties allow further investigation in the mutant MEK setting upon BRAF/MEK therapy.


Subject(s)
Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Adenosine Triphosphate/metabolism , Cell Line, Tumor , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics
2.
J Transl Autoimmun ; 4: 100103, 2021.
Article in English | MEDLINE | ID: mdl-34041473

ABSTRACT

Impaired interleukin-2 (IL-2) production and regulatory T-cell dysfunction have been implicated as immunological mechanisms central to the pathogenesis of multiple autoimmune and inflammatory diseases. NKTR-358, a novel regulatory T-cell stimulator, is an investigational therapeutic that selectively restores regulatory T-cell homeostasis in these diseases. We investigated NKTR-358's selectivity for regulatory T-cells, receptor-binding properties, ex vivo and in vivo pharmacodynamics, ability to suppress conventional T-cell proliferation in mice and non-human primates, and functional activity in a murine model of systemic lupus erythematosus. In vitro, NKTR-358 demonstrated decreased affinity for IL-2Rα, IL-2Rß, and IL-2Rαß compared with recombinant human IL-2 (rhIL-2). A single dose of NKTR-358 in cynomolgus monkeys produced a greater than 15-fold increase in regulatory T-cells, and the increase lasted until day 14, while daily rhIL-2 administration for 5 days only elicited a 3-fold increase, which lasted until day 7. Repeated dosing of NKTR-358 over 6 months in cynomolgus monkeys elicited cyclical, robust increases in regulatory T-cells with no loss in drug activity over the course of treatment. Regulatory T-cells isolated from NKTR-358-treated mice displayed a sustained, higher suppression of conventional T-cell proliferation than regulatory T-cells isolated from vehicle-treated mice. NKTR-358 treatment in a mouse model (MRL/MpJ-Faslpr) of systemic lupus erythematosus for 12 weeks maintained elevated regulatory T-cells for the treatment duration and ameliorated disease progression. Together, these results suggest that NKTR-358 has the ability to elicit sustained and preferential proliferation and activation of regulatory T-cells without corresponding effects on conventional T-cells, with improved pharmacokinetics compared with rhIL-2.

3.
J Med Chem ; 63(23): 14885-14904, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33258605

ABSTRACT

Overexpression of PIM 1, 2, and 3 kinases is frequently observed in many malignancies. Previously, we discovered a potent and selective pan-PIM kinase inhibitor, compound 2, currently in phase I clinical trials. In this work, we were interested in replacing the amino group on the cyclohexane ring in compound 2 with a hydroxyl group. Structure-based drug design led to cellularly potent but metabolically unstable tetra-substituted cyclohexyl diols. Efforts on the reduction of Log D by introducing polar heterocycles improved metabolic stability. Incorporating fluorine to the tetra-substituted cyclohexyl diol moiety further reduced Log D, resulting in compound 14, a cellularly potent tetra-substituted cyclohexyl diol inhibitor with moderate metabolic stability and good permeability. We also describe the development of efficient and scalable synthetic routes toward synthetically challenging tetra-substituted cyclohexyl diol compounds. In particular, intermediate 36 was identified as a versatile intermediate, enabling a large-scale synthesis of highly substituted cyclohexane derivatives.


Subject(s)
Cyclohexanols/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Cell Line, Tumor , Cyclohexanols/chemical synthesis , Cyclohexanols/metabolism , Humans , Microsomes, Liver/metabolism , Molecular Docking Simulation , Molecular Structure , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Structure-Activity Relationship
4.
PLoS One ; 12(7): e0179431, 2017.
Article in English | MEDLINE | ID: mdl-28678791

ABSTRACT

Cytokines are potent immune modulating agents but are not ideal medicines in their natural form due to their short half-life and pleiotropic systemic effects. NKTR-214 is a clinical-stage biologic that comprises interleukin-2 (IL2) protein bound by multiple releasable polyethylene glycol (PEG) chains. In this highly PEG-bound form, the IL2 is inactive; therefore, NKTR-214 is a biologic prodrug. When administered in vivo, the PEG chains slowly release, creating a cascade of increasingly active IL2 protein conjugates bound by fewer PEG chains. The 1-PEG-IL2 and 2-PEG-IL2 species derived from NKTR-214 are the most active conjugated-IL2 species. Free-IL2 protein is undetectable in vivo as it is eliminated faster than formed. The PEG chains on NKTR-214 are located at the region of IL2 that contacts the alpha (α) subunit of the heterotrimeric IL2 receptor complex, IL2Rαßγ, reducing its ability to bind and activate the heterotrimer. The IL2Rαßγ complex is constitutively expressed on regulatory T cells (Tregs). Therefore, without the use of mutations, PEGylation reduces the affinity for IL2Rαßγ to a greater extent than for IL2Rßγ, the receptor complex predominant on CD8 T cells. NKTR-214 treatment in vivo favors activation of CD8 T cells over Tregs in the tumor microenvironment to provide anti-tumor efficacy in multiple syngeneic models. Mechanistic modeling based on in vitro and in vivo kinetic data provides insight into the mechanism of NKTR-214 pharmacology. The model reveals that conjugated-IL2 protein derived from NKTR-214 occupy IL-2Rßγ to a greater extent compared to free-IL2 protein. The model accurately describes the sustained in vivo signaling observed after a single dose of NKTR-214 and explains how the properties of NKTR-214 impart a unique kinetically-controlled immunological mechanism of action.


Subject(s)
Immunotherapy/methods , Interleukin-2/analogs & derivatives , Neoplasms/therapy , Polyethylene Glycols/pharmacology , Receptors, Interleukin-2/agonists , Algorithms , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Drug Liberation , Female , Interleukin Receptor Common gamma Subunit/agonists , Interleukin Receptor Common gamma Subunit/metabolism , Interleukin-2/pharmacokinetics , Interleukin-2/pharmacology , Interleukin-2 Receptor alpha Subunit/agonists , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-2 Receptor beta Subunit/agonists , Interleukin-2 Receptor beta Subunit/metabolism , Kinetics , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Models, Theoretical , Neoplasms/immunology , Neoplasms/metabolism , Phosphorylation/drug effects , Polyethylene Glycols/pharmacokinetics , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Receptors, Interleukin-2/metabolism , STAT5 Transcription Factor/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transplantation, Homologous , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
5.
Clin Cancer Res ; 22(3): 680-90, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26832745

ABSTRACT

PURPOSE: Aldesleukin, recombinant human IL2, is an effective immunotherapy for metastatic melanoma and renal cancer, with durable responses in approximately 10% of patients; however, severe side effects limit maximal dosing and thus the number of patients able to receive treatment and potential cure. NKTR-214 is a prodrug of conjugated IL2, retaining the same amino acid sequence as aldesleukin. The IL2 core is conjugated to 6 releasable polyethylene glycol (PEG) chains. In vivo, the PEG chains slowly release to generate active IL2 conjugates. EXPERIMENTAL DESIGN: We evaluated the bioactivity and receptor binding of NKTR-214 and its active IL2 conjugates in vitro; the tumor immunology, tumor pharmacokinetics, and efficacy of NKTR-214 as a single agent and in combination with anti-CTLA-4 antibody in murine tumor models. Tolerability was evaluated in non-human primates. RESULTS: In a murine melanoma tumor model, the ratio of tumor-killing CD8(+) T cells to Foxp3(+) regulatory T cells was greater than 400 for NKTR-214 compared with 18 for aldesleukin, supporting preferential activation of the IL2 receptor beta over IL2 receptor alpha, due to the location of PEG molecules. NKTR-214 provides a 500-fold greater exposure of the tumor to conjugated IL2 compared with aldesleukin. NKTR-214 showed efficacy as a single agent and provided durable immunity that was resistant to tumor rechallenge in combination with anti-CTLA-4 antibody. NKTR-214 was well tolerated in non-human primates. CONCLUSIONS: These data support further evaluation of NKTR-214 in humans for a variety of tumor types, adding to the repertoire of potent and potentially curative cancer immunotherapies.


Subject(s)
Antineoplastic Agents/pharmacology , Interleukin-2/analogs & derivatives , Neoplasms/metabolism , Neoplasms/pathology , Polyethylene Glycols/pharmacology , Prodrugs , Receptors, Interleukin-2/metabolism , Recombinant Fusion Proteins/pharmacology , Animals , Antineoplastic Agents/chemistry , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CTLA-4 Antigen/antagonists & inhibitors , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , Female , Humans , Immunologic Memory , Interleukin-2/chemistry , Interleukin-2/pharmacology , Lymphocytes, Tumor-Infiltrating , Male , Melanoma, Experimental , Mice , Models, Molecular , Molecular Conformation , Neoplasms/drug therapy , Neoplasms/immunology , Polyethylene Glycols/chemistry , Protein Binding , Receptors, Interleukin-2/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Proteins/pharmacology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Burden/drug effects
6.
J Med Chem ; 58(21): 8373-86, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26505898

ABSTRACT

Pan proviral insertion site of Moloney murine leukemia (PIM) 1, 2, and 3 kinase inhibitors have recently begun to be tested in humans to assess whether pan PIM kinase inhibition may provide benefit to cancer patients. Herein, the synthesis, in vitro activity, in vivo activity in an acute myeloid leukemia xenograft model, and preclinical profile of the potent and selective pan PIM kinase inhibitor compound 8 (PIM447) are described. Starting from the reported aminopiperidyl pan PIM kinase inhibitor compound 3, a strategy to improve the microsomal stability was pursued resulting in the identification of potent aminocyclohexyl pan PIM inhibitors with high metabolic stability. From this aminocyclohexyl series, compound 8 entered the clinic in 2012 in multiple myeloma patients and is currently in several phase 1 trials of cancer patients with hematological malignancies.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Picolinic Acids/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Amides/chemical synthesis , Amides/chemistry , Amides/therapeutic use , Animals , Cell Line, Tumor , Halogenation , Humans , Leukemia, Myeloid, Acute/metabolism , Mice , Models, Molecular , Picolinic Acids/chemical synthesis , Picolinic Acids/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism
7.
Clin Cancer Res ; 20(7): 1834-45, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24474669

ABSTRACT

PURPOSE: PIM kinases have been shown to act as oncogenes in mice, with each family member being able to drive progression of hematologic cancers. Consistent with this, we found that PIMs are highly expressed in human hematologic cancers and show that each isoform has a distinct expression pattern among disease subtypes. This suggests that inhibitors of all three PIMs would be effective in treating multiple hematologic malignancies. EXPERIMENTAL DESIGN: Pan-PIM inhibitors have proven difficult to develop because PIM2 has a low Km for ATP and, thus, requires a very potent inhibitor to effectively block the kinase activity at the ATP levels in cells. We developed a potent and specific pan-PIM inhibitor, LGB321, which is active on PIM2 in the cellular context. RESULTS: LGB321 is active on PIM2-dependent multiple myeloma cell lines, where it inhibits proliferation, mTOR-C1 signaling and phosphorylation of BAD. Broad cancer cell line profiling of LGB321 demonstrates limited activity in cell lines derived from solid tumors. In contrast, significant activity in cell lines derived from diverse hematological lineages was observed, including acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), multiple myeloma and non-Hodgkin lymphoma (NHL). Furthermore, we demonstrate LGB321 activity in the KG-1 AML xenograft model, in which modulation of pharmacodynamics markers is predictive of efficacy. Finally, we demonstrate that LGB321 synergizes with cytarabine in this model. CONCLUSIONS: We have developed a potent and selective pan-PIM inhibitor with single-agent antiproliferative activity and show that it synergizes with cytarabine in an AML xenograft model. Our results strongly support the development of Pan-PIM inhibitors to treat hematologic malignancies.


Subject(s)
Hematologic Neoplasms/therapy , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins/genetics , Animals , Cell Line, Tumor , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Humans , Mice , Phosphorylation , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics , Xenograft Model Antitumor Assays
8.
Blood ; 122(9): 1610-20, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23818547

ABSTRACT

Multiple myeloma (MM) is the second most common hematologic malignancy. Despite recent treatment advances, it remains incurable. Here, we report that Pim2 kinase expression is highly elevated in MM cells and demonstrate that it is required for MM cell proliferation. Functional interference of Pim2 activity either by short hairpin RNAs or by a potent and selective small-molecule inhibitor leads to significant inhibition of MM cell proliferation. Pim inhibition results in a significant decrease of mammalian target of rapamycin C1 (mTOR-C1) activity, which is critical for cell proliferation. We identify TSC2, a negative regulator of mTOR-C1, as a novel Pim2 substrate and show that Pim2 directly phosphorylates TSC2 on Ser-1798 and relieves the suppression of TSC2 on mTOR-C1. These findings support Pim2 as a promising therapeutic target for MM and define a novel Pim2-TSC2-mTOR-C1 pathway that drives MM proliferation.


Subject(s)
Cell Proliferation , Multiple Myeloma/pathology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Tumor Suppressor Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Models, Biological , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Multiprotein Complexes/metabolism , Phosphorylation/drug effects , Phosphorylation/genetics , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Pyridines/pharmacology , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 2 Protein , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays
9.
ACS Med Chem Lett ; 4(12): 1193-7, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24900629

ABSTRACT

Proviral insertion of Moloney virus (PIM) 1, 2, and 3 kinases are serine/threonine kinases that normally function in survival and proliferation of hematopoietic cells. As high expression of PIM1, 2, and 3 is frequently observed in many human malignancies, including multiple myeloma, non-Hodgkins lymphoma, and myeloid leukemias, there is interest in determining whether selective PIM inhibition can improve outcomes of these human cancers. Herein, we describe our efforts toward this goal. The structure guided optimization of a singleton high throughput screening hit in which the potency against all three PIM isoforms was increased >10,000-fold to yield compounds with pan PIM K is < 10 pM, nanomolar cellular potency, and in vivo activity in an acute myeloid leukemia Pim-dependent tumor model is described.

10.
Cancer Cell ; 20(6): 781-96, 2011 Dec 13.
Article in English | MEDLINE | ID: mdl-22172723

ABSTRACT

Tumor immune surveillance and cancer immunotherapies are thought to depend on the intratumoral infiltration of activated CD8(+) T cells. Intratumoral CD8(+) T cells are rare and lack activity. IL-10 is thought to contribute to the underlying immune suppressive microenvironment. Defying those expectations we demonstrate that IL-10 induces several essential mechanisms for effective antitumor immune surveillance: infiltration and activation of intratumoral tumor-specific cytotoxic CD8(+) T cells, expression of the Th1 cytokine interferon-γ (IFNγ) and granzymes in CD8(+) T cells, and intratumoral antigen presentation molecules. Consequently, tumor immune surveillance is weakened in mice deficient for IL-10 whereas transgenic overexpression of IL-10 protects mice from carcinogenesis. Treatment with pegylated IL-10 restores tumor-specific intratumoral CD8(+) T cell function and controls tumor growth.


Subject(s)
Interferon-gamma/metabolism , Interleukin-10/metabolism , Neoplasms, Experimental/immunology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/metabolism , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic , Female , Granzymes/metabolism , Humans , Interferon-gamma/genetics , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-10 Receptor alpha Subunit/genetics , Interleukin-10 Receptor alpha Subunit/metabolism , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Perforin/metabolism , Skin Neoplasms/chemically induced , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Spleen/metabolism , Transplantation, Heterologous , Tumor Burden , Tumor Escape
11.
Mol Cell Endocrinol ; 280(1-2): 6-12, 2008 Jan 02.
Article in English | MEDLINE | ID: mdl-17945411

ABSTRACT

Epidermal gamma delta T cells (gammadeltaT) and Langerhans cells (LC) are immune cells altered by exposure to ultraviolet radiation (UVB), a powerful stressor resulting in immune suppression. Prolactin (PRL) has been characterized as an immunomodulator, particularly during stress. In this study, we have asked whether separate administration of the 2 major forms of prolactin, unmodified and phosphorylated, to groups of 15 mice (3 experiments, each with 5 mice per treatment group) affected the number and morphology of these epidermal immune cells under control conditions, and following UV-irradiation. Under control conditions, both PRLs reduced the number of gammadeltaT, but a molecular mimic of phosphorylated PRL (S179D PRL) was more effective, resulting in a 30% reduction. In the irradiated group, however, S179D PRL was protective against a UV-induced reduction in gammadeltaT number and change in morphology (halved the reduction and normalized the morphology). In addition, S179D PRL, but not unmodified (U-PRL), maintained a normal LC:gammadeltaT ratio and sustained the dendritic morphology of LC after UV exposure. These findings suggest that S179D PRL may have an overall protective effect, countering UV-induced cellular alterations in the epidermis.


Subject(s)
Epidermis/drug effects , Immunologic Factors/pharmacology , Langerhans Cells/drug effects , Prolactin/pharmacology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes/drug effects , Ultraviolet Rays , Animals , Epidermis/metabolism , Epidermis/radiation effects , Immunologic Factors/administration & dosage , Immunologic Factors/metabolism , Infusion Pumps, Implantable , Langerhans Cells/metabolism , Langerhans Cells/radiation effects , Male , Mice , Mice, Inbred C3H , Phosphorylation , Prolactin/administration & dosage , Prolactin/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/radiation effects
12.
Trends Immunol ; 28(5): 207-12, 2007 May.
Article in English | MEDLINE | ID: mdl-17395538

ABSTRACT

During tumorigenesis, selective pressure drives tumor cells to develop several strategies that enable growth and propagation. Transformed cells produce or elicit factors that provide growth signals, nutrients and a favorable tumor microenvironment. In addition, tumor cells can evade elimination by the immune system by several mechanisms, including developing resistance to T cell-induced apoptosis or the local expression of immune-modulatory molecules and cytokines. Recently, we described a role for the cytokine interleukin (IL)-23 in promoting tumor incidence and growth. In addition, IL-23 not only stimulates neutrophil and macrophage infiltration, but also promotes angiogenesis and inflammatory mediators in the tumor microenvironment. IL-23 antagonizes IL-12 and interferon gamma, both of which are essential cytokines for cytotoxic immune responses, and controls the influx and activity of anti-tumor effector lymphocytes. We suggest that IL-23 inflicts a repurposing of the adaptive cytotoxic effector response away from anti-tumor immunity ('sword') and towards proinflammatory and proangiogenic effector pathways that nourish the tumor ('plowshare'). Consequently, IL-23 enables the persistence of the recognized tumor cells, accompanied by tumor-associated inflammation. This concept can explain tumor growth in the presence of large quantities of tumor-specific T cells.


Subject(s)
Immunologic Surveillance , Inflammation/pathology , Interleukin-23/immunology , Neoplasms/immunology , Neoplasms/pathology , Animals , CD8-Positive T-Lymphocytes/immunology , Humans , Interleukin-17/metabolism , Interleukin-23/metabolism , Mice , Neoplasms/blood supply , Neoplasms/metabolism , Neovascularization, Pathologic/immunology
13.
Nature ; 442(7101): 461-5, 2006 Jul 27.
Article in English | MEDLINE | ID: mdl-16688182

ABSTRACT

Chronic inflammation has long been associated with increased incidence of malignancy and similarities in the regulatory mechanisms have been suggested for more than a century. Infiltration of innate immune cells, elevated activities of matrix metalloproteases and increased angiogenesis and vasculature density are a few examples of the similarities between chronic and tumour-associated inflammation. Conversely, the elimination of early malignant lesions by immune surveillance, which relies on the cytotoxic activity of tumour-infiltrating T cells or intra-epithelial lymphocytes, is thought to be rate-limiting for the risk to develop cancer. Here we show a molecular connection between the rise in tumour-associated inflammation and a lack of tumour immune surveillance. Expression of the heterodimeric cytokine interleukin (IL)-23, but not of its close relative IL-12, is increased in human tumours. Expression of these cytokines antagonistically regulates local inflammatory responses in the tumour microenvironment and infiltration of intra-epithelial lymphocytes. Whereas IL-12 promotes infiltration of cytotoxic T cells, IL-23 promotes inflammatory responses such as upregulation of the matrix metalloprotease MMP9, and increases angiogenesis but reduces CD8 T-cell infiltration. Genetic deletion or antibody-mediated elimination of IL-23 leads to increased infiltration of cytotoxic T cells into the transformed tissue, rendering a protective effect against chemically induced carcinogenesis. Finally, transplanted tumours are growth-restricted in hosts depleted for IL-23 or in IL-23-receptor-deficient mice. Although many strategies for immune therapy of cancer attempt to stimulate an immune response against solid tumours, infiltration of effector cells into the tumour tissue often appears to be a critical hurdle. We show that IL-23 is an important molecular link between tumour-promoting pro-inflammatory processes and the failure of the adaptive immune surveillance to infiltrate tumours.


Subject(s)
Interleukins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Division , Genetic Predisposition to Disease , Humans , Inflammation/genetics , Inflammation/pathology , Interleukin-12/biosynthesis , Interleukin-12/immunology , Interleukin-23 , Interleukin-23 Subunit p19 , Interleukins/biosynthesis , Interleukins/deficiency , Interleukins/genetics , Mice , Mice, Inbred C57BL , Neoplasms/genetics , Neoplasms/immunology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...