Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Mol Med (Berl) ; 98(1): 135-148, 2020 01.
Article in English | MEDLINE | ID: mdl-31838577

ABSTRACT

The B7 family member, B7H6, is a ligand for the natural killer cell receptor NKp30. B7H6 is hardly expressed on normal tissues, but undergoes upregulation on different types of tumors, implicating it as an attractive target for cancer immunotherapy. The molecular mechanisms that control B7H6 expression are poorly understood. We report that in contrast to other NK cell ligands, endoplasmic reticulum (ER) stress upregulates B7H6 mRNA levels and surface expression. B7H6 induction by ER stress requires protein kinase R-like ER kinase (PERK), one of the three canonical sensors of the unfolded protein response. PERK phosphorylates eIF2α, which regulates protein synthesis and gene expression. Because eIF2α is phosphorylated by several kinases following different stress conditions, the program downstream to eIF2α phosphorylation is called the integrated stress response (ISR). Several drugs were reported to promote the ISR. Nelfinavir and lopinavir, two clinically approved HIV protease inhibitors, promote eIF2α phosphorylation by different mechanisms. We show that nelfinavir and lopinavir sustainably instigate B7H6 expression at their pharmacologically relevant concentrations. As such, ER stress and ISR conditions sensitize melanoma targets to CAR-T cells directed against B7H6. Our study highlights a novel mechanism to induce B7H6 expression and suggests a pharmacological approach to improve B7H6-directed immunotherapy. KEY MESSAGES: B7H6 is induced by ER stress in a PERK-dependent mechanism. Induction of B7H6 is obtained pharmacologically by HIV protease inhibitors. Exposure of tumor cells to the HIV protease inhibitor nelfinavir improves the recognition by B7H6-directed CAR-T.


Subject(s)
B7 Antigens/metabolism , Endoplasmic Reticulum Stress/genetics , Eukaryotic Initiation Factor-2/metabolism , HIV Protease Inhibitors/pharmacology , Lopinavir/pharmacology , Nelfinavir/pharmacology , Signal Transduction/drug effects , B7 Antigens/genetics , Blood Donors , Cell Line, Tumor , Humans , Immunotherapy, Adoptive/methods , Killer Cells, Natural/immunology , Phosphorylation/drug effects , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/immunology , Transduction, Genetic , Transfection , Unfolded Protein Response/drug effects , Unfolded Protein Response/genetics , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
2.
BioDrugs ; 33(5): 515-537, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31363930

ABSTRACT

Chimeric antigen receptor-T cells (CAR-Ts) are an exciting new cancer treatment modality exemplified by the recent regulatory approval of two CD19-targeted CAR-T therapies for certain B cell malignancies. However, this success in the hematological setting has yet to translate to a significant level of objective clinical responses in the solid tumor setting. The reason for this lack of translation undoubtedly lies in the substantial challenges raised by solid tumors to all therapies, including CAR-T, that differ from B cell malignancies. For instance, intravenously infused CAR-Ts are likely to make rapid contact with cancerous B cells since both tend to reside in the same vascular compartments within the body. By contrast, solid cancers tend to form discrete tumor masses with an immune-suppressive tumor microenvironment composed of tumor cells and non-tumor stromal cells served by abnormal vasculature that restricts lymphocyte infiltration and suppresses immune function, expansion, and persistence. Moreover, the paucity of uniquely and homogeneously expressed tumor antigens and inherent plasticity of cancer cells provide major challenges to the specificity, potency, and overall effectiveness of CAR-T therapies. This review focuses on the major preclinical and clinical strategies currently being pursued to tackle these challenges in order to drive the success of CAR-T therapy against solid tumors.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Neoplasms/therapy , Receptors, Chimeric Antigen/therapeutic use , Tumor Microenvironment/immunology , Animals , Cell- and Tissue-Based Therapy/adverse effects , Clinical Trials as Topic , Humans , Neoplasms/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation
3.
Front Immunol ; 9: 2940, 2018.
Article in English | MEDLINE | ID: mdl-30619300

ABSTRACT

Chimeric Antigen Receptor (CAR) T cells expressing the fusion of the NKG2D protein with CD3ζ (NKG2D-CAR T Cells) acquire a specificity for stress-induced ligands expressed on hematological and solid cancers. However, these stress ligands are also transiently expressed by activated T cells implying that NKG2D-based T cells may undergo self-killing (fratricide) during cell manufacturing or during the freeze thaw cycle prior to infusion in patients. To avoid target-driven fratricide and enable the production of NKG2D-CAR T cells for clinical application, two distinct approaches were investigated. The first focused upon the inclusion of a Phosphoinositol-3-Kinase inhibitor (LY294002) into the production process. A second strategy involved the inclusion of antibody blockade of NKG2D itself. Both processes impacted T cell fratricide, albeit at different levels with the antibody process being the most effective in terms of cell yield. While both approaches generated comparable NKG2D-CAR T cells, there were subtle differences, for example in differentiation status, that were fine-tuned through the phasing of the inhibitor and antibody during culture in order to generate a highly potent NKG2D-CAR T cell product. By means of targeted inhibition of NKG2D expression or generic inhibition of enzyme function, target-driven CAR T fratricide can be overcome. These strategies have been incorporated into on-going clinical trials to enable a highly efficient and reproducible manufacturing process for NKG2D-CAR T cells.


Subject(s)
Cytotoxicity, Immunologic/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Antibodies, Blocking/immunology , Antibodies, Blocking/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Chromones/pharmacology , Cytotoxicity, Immunologic/drug effects , Enzyme Inhibitors/pharmacology , Humans , Immunotherapy, Adoptive/methods , K562 Cells , Ligands , Morpholines/pharmacology , NK Cell Lectin-Like Receptor Subfamily K/antagonists & inhibitors , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism
4.
Cell Mol Life Sci ; 73(24): 4739-4748, 2016 12.
Article in English | MEDLINE | ID: mdl-27436342

ABSTRACT

Genomic instability drives cancer progression by promoting genetic abnormalities that allow for the multi-step clonal selection of cells with growth advantages. We previously reported that the IL-9-dependent TS1 cell line sequentially acquired activating substitutions in JAK1 and JAK3 upon successive selections for growth factor independent and JAK inhibitor-resistant cells, suggestive of a defect in mutation avoidance mechanisms. In the first part of this paper, we discovered that the gene encoding mutL homolog-1 (MLH1), a key component of the DNA mismatch repair system, is silenced by promoter methylation in TS1 cells. By means of stable ectopic expression and RNA interference methods, we showed that the high frequencies of growth factor-independent and inhibitor-resistant cells with activating JAK mutations can be attributed to the absence of MLH1 expression. In the second part of this paper, we confirm the clinical relevance of our findings by showing that chronic myeloid leukemia relapses upon ABL-targeted therapy correlated with a lower expression of MLH1 messenger RNA. Interestingly, the mutational profile observed in our TS1 model, characterized by a strong predominance of T:A>C:G transitions, was identical to the one described in the literature for primitive cells derived from chronic myeloid leukemia patients. Taken together, our observations demonstrate for the first time a causal relationship between MLH1-deficiency and incidence of oncogenic point mutations in tyrosine kinases driving cell transformation and acquired resistance to kinase-targeted cancer therapies.


Subject(s)
Drug Resistance, Neoplasm/genetics , Janus Kinases/genetics , MutL Protein Homolog 1/metabolism , Oncogenes , Point Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Animals , Cell Line , Clone Cells , DNA Methylation/drug effects , DNA Methylation/genetics , Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Leukemic/drug effects , Gene Knockdown Techniques , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , MutL Protein Homolog 1/genetics , Promoter Regions, Genetic/genetics , RNA, Small Interfering/metabolism
5.
Haematologica ; 100(10): 1240-53, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26432382

ABSTRACT

Constitutive JAK-STAT pathway activation occurs in most myeloproliferative neoplasms as well as in a significant proportion of other hematologic malignancies, and is frequently a marker of poor prognosis. The underlying molecular alterations are heterogeneous as they include activating mutations in distinct components (cytokine receptor, JAK, STAT), overexpression (cytokine receptor, JAK) or rare JAK2 fusion proteins. In some cases, concomitant loss of negative regulators contributes to pathogenesis by further boosting the activation of the cascade. Exploiting the signaling bottleneck provided by the limited number of JAK kinases is an attractive therapeutic strategy for hematologic neoplasms driven by constitutive JAK-STAT pathway activation. However, given the conserved nature of the kinase domain among family members and the interrelated roles of JAK kinases in many physiological processes, including hematopoiesis and immunity, broad usage of JAK inhibitors in hematology is challenged by their narrow therapeutic window. Novel therapies are, therefore, needed. The development of more selective inhibitors is a questionable strategy as such inhibitors might abrogate the beneficial contribution of alleviating the cancer-related pro-inflammatory microenvironment and raise selective pressure to a threshold that allows the emergence of malignant subclones harboring drug-resistant mutations. In contrast, synergistic combinations of JAK inhibitors with drugs targeting cascades that work in concert with JAK-STAT pathway appear to be promising therapeutic alternatives to JAK inhibitors as monotherapies.


Subject(s)
Antineoplastic Agents/therapeutic use , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/metabolism , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Molecular Targeted Therapy , Protein Kinase Inhibitors/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Genetic Variation , Hematologic Neoplasms/genetics , Hematopoiesis/genetics , Humans , Janus Kinases/genetics , Mutation , Protein Kinase Inhibitors/pharmacology , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction/drug effects
6.
J Biol Chem ; 290(48): 29022-34, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26446793

ABSTRACT

JAK1 and JAK3 are recurrently mutated in acute lymphoblastic leukemia. These tyrosine kinases associate with heterodimeric cytokine receptors such as IL-7 receptor or IL-9 receptor, in which JAK1 is appended to the specific chain, and JAK3 is appended to the common gamma chain. Here, we studied the role of these receptor complexes in mediating the oncogenic activity of JAK3 mutants. Although JAK3(V674A) and the majority of other JAK3 mutants needed to bind to a functional cytokine receptor complex to constitutively activate STAT5, JAK3(L857P) was unexpectedly found to not depend on such receptor complexes for its activity, which was induced without receptor or JAK1 co-expression. Introducing a mutation in the FERM domain that abolished JAK-receptor interaction did not affect JAK3(L857P) activity, whereas it inhibited the other receptor-dependent mutants. The same cytokine receptor independence as for JAK3(L857P) was observed for homologous Leu(857) mutations of JAK1 and JAK2 and for JAK3(L875H). This different cytokine receptor requirement correlated with different functional properties in vivo and with distinct sensitivity to JAK inhibitors. Transduction of murine hematopoietic cells with JAK3(V674A) led homogenously to lymphoblastic leukemias in BALB/c mice. In contrast, transduction with JAK3(L857P) induced various types of lymphoid and myeloid leukemias. Moreover, ruxolitinib, which preferentially blocks JAK1 and JAK2, abolished the proliferation of cells transformed by the receptor-dependent JAK3(V674A), yet proved much less potent on cells expressing JAK3(L857P). These particular cells were, in contrast, more sensitive to JAK3-specific inhibitors. Altogether, our results showed that different JAK3 mutations induce constitutive activation through distinct mechanisms, pointing to specific therapeutic perspectives.


Subject(s)
Janus Kinase 3 , Mutation, Missense , Protein Kinase Inhibitors/pharmacology , Amino Acid Substitution , Animals , Cell Line, Tumor , Humans , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/genetics , Janus Kinase 3/metabolism , Mice , Mice, Inbred BALB C , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism
7.
Blood ; 124(26): 3924-31, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25352124

ABSTRACT

The acquisition of growth signal self-sufficiency is 1 of the hallmarks of cancer. We previously reported that the murine interleukin-9-dependent TS1 cell line gives rise to growth factor-independent clones with constitutive activation of the Janus kinase (JAK)- signal transducer and activator of transcription (STAT) pathway. Here, we show that this transforming event results from activating mutations either in JAK1, JAK3, or in both kinases. Transient and stable expression of JAK1 and/or JAK3 mutants showed that each mutant induces STAT activation and that their coexpression further increases this activation. The proliferation of growth factor-independent TS1 clones can be efficiently blocked by JAK inhibitors such as ruxolitinib or CMP6 in short-term assays. However, resistant clones occur upon long-term culture in the presence of inhibitors. Surprisingly, resistance to CMP6 was not caused by the acquisition of secondary mutations in the adenosine triphosphate-binding pocket of the JAK mutant. Indeed, cells that originally showed a JAK1-activating mutation became resistant to inhibitors by acquiring another activating mutation in JAK3, whereas cells that originally showed a JAK3-activating mutation became resistant to inhibitors by acquiring another activating mutation in JAK1. These observations underline the cooperation between JAK1 and JAK3 mutants in T-cell transformation and represent a new mechanism of acquisition of resistance against JAK inhibitors.


Subject(s)
Drug Resistance, Neoplasm , Janus Kinase 1/genetics , Janus Kinase 3/genetics , Protein Kinase Inhibitors/chemistry , Adenosine Triphosphate/chemistry , Animals , Cell Line , Cell Proliferation , Cell Transformation, Neoplastic , HEK293 Cells , Humans , Janus Kinases/antagonists & inhibitors , Mice , Mutation, Missense , Nitriles , Point Mutation , Protein Structure, Tertiary , Pyrazoles/chemistry , Pyrimidines , Signal Transduction
8.
Haematologica ; 96(6): 845-53, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21393331

ABSTRACT

BACKGROUND: Activating mutations in JAK1 and JAK2 have been described in patients with various hematologic malignancies including acute lymphoblastic leukemia and myeloproliferative neoplasms, leading to clinical trials with JAK inhibitors. While there has been a tremendous effort towards the development of specific JAK inhibitors, mutations conferring resistance to such drugs have not yet been observed. DESIGN AND METHODS: Taking advantage of a model of spontaneous cellular transformation, we sequenced JAK1 in selected tumorigenic BaF3 clones and identified 25 de novo JAK1 activating mutations, including 5 mutations already described in human leukemias. We further used this library of JAK1 mutation-positive cell lines to assess their sensitivity to ATP-competitive inhibitors. RESULTS: While most JAK1 mutants were sensitive to ATP-competitive JAK inhibitors, mutations targeting Phe958 and Pro960 in the hinge region of the kinase domain rendered JAK1 constitutively active but also resistant to all tested JAK inhibitors. Furthermore, mutation of the homologous Tyr931 in JAK2 wild-type or JAK2 V617F mutant found in patients with myeloproliferative neoplasms also conferred resistance to JAK inhibitors, such as INCB018424, which is currently in clinical use. CONCLUSIONS: Our data indicate that some activating mutations not only promote autonomous cell proliferation but also confer resistance to ATP-competitive inhibitors. In vivo, such a mutation can potentially occur as primary JAK-activating mutations but also as secondary mutations combining oncogenicity with drug resistance.


Subject(s)
Adenosine Triphosphate/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , Janus Kinase 1/genetics , Janus Kinase 2/genetics , Mutation/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/physiopathology , Animals , Binding, Competitive/drug effects , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Gene Order , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Mice , Models, Molecular , Phosphorylation/drug effects , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Structure, Secondary , STAT5 Transcription Factor/antagonists & inhibitors , STAT5 Transcription Factor/metabolism , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...