71 results on '"Sloma I"'
Search Results
2. Topic: AS06-Prognosis/AS06a-Prognostic factors of outcome and risk assessment: TET2 MUTATIONAL STATUS AFFECTS MYELODYSPLASTIC SYNDROME EVOLUTION TO CHRONIC MYELOMONOCYTIC LEUKEMIA
- Author
-
Quang, V. Tran, primary, Podvin, B., additional, Desterke, C., additional, Tarfi, S., additional, Barathon, Q., additional, Bouchra, B., additional, Freynet, N., additional, Parinet, V., additional, Leclerc, M., additional, Sébastien, M., additional, Solary, E., additional, Selimoglu-Buet, D., additional, Duployez, N., additional, Wagner-Ballon, O., additional, and Sloma, I., additional
- Published
- 2023
- Full Text
- View/download PDF
3. Leucémie myéloïde chronique des sujets jeunes : expérience dans une unité d’hématologie clinique au Sénégal
- Author
-
Fall, S., Ndiaye, F.S.D., Dior, O.D., Sall, A.F., Sloma, I., Touré-Fall, A., and Keita, Y.
- Published
- 2015
- Full Text
- View/download PDF
4. P098 - Topic: AS06-Prognosis/AS06a-Prognostic factors of outcome and risk assessment: TET2 MUTATIONAL STATUS AFFECTS MYELODYSPLASTIC SYNDROME EVOLUTION TO CHRONIC MYELOMONOCYTIC LEUKEMIA
- Author
-
Quang, V. Tran, Podvin, B., Desterke, C., Tarfi, S., Barathon, Q., Bouchra, B., Freynet, N., Parinet, V., Leclerc, M., Sébastien, M., Solary, E., Selimoglu-Buet, D., Duployez, N., Wagner-Ballon, O., and Sloma, I.
- Published
- 2023
- Full Text
- View/download PDF
5. Insights into the stem cells of chronic myeloid leukemia
- Author
-
Sloma, I, Jiang, X, Eaves, A C, and Eaves, C J
- Published
- 2010
- Full Text
- View/download PDF
6. CLONAL ARCHITECTURE OF RELAPSED OR REFRACTORY FOLLICULAR HELPER T‐CELL LYMPHOMA: AN ANCILLARY STUDY OF THE ORACLE TRIAL, A LYSA STUDY.
- Author
-
Loyaux, R., Sako, N., Quang, V. Tran, Bachy, E., Morschhauser, F., Cartron, G., Gressin, R., Daguindau, N., Le Gouill, S., Wolfromm, A., Bouabdallah, K., Ysebaert, L., Casasnovas, O., Robe, C., Delfau, M., Dupuis, J., De Leval, L., Gaulard, P., Sloma, I., and Lemonnier, F.
- Subjects
T-cell lymphoma - Abstract
Demonstration of I TET2 i and I DNMT3A i mutations in B cells, myeloid cells, or hematopoietic progenitor cells have suggested that TFHL can emerge from clonal hematopoiesis (CH) in a multi-step process. Follicular helper T-cell lymphoma (TFHL) results from the oncogenic transformation of a TFH cell, driven by mutations in genes involved in epigenetic regulation ( I TET2 i , I DNMT3A, IDH2 i ) and T-cell signaling ( I RHOA i ). Meanwhile, bulk BM cells from 29 patients were sequenced with the same NGS panel and then compared to the mutations found in cfDNA (31 patients) and tumor biopsies (28 patients). [Extracted from the article]
- Published
- 2023
- Full Text
- View/download PDF
7. Ex vivo expansion of normal and chronic myeloid leukemic stem cells without functional alteration using a NUP98HOXA10homeodomain fusion gene
- Author
-
Sloma, I, Imren, S, Beer, P A, Zhao, Y, Lecault, V, Leung, D, Raghuram, K, Brimacombe, C, Lambie, K, Piret, J, Hansen, C, Humphries, R K, and Eaves, C J
- Published
- 2013
- Full Text
- View/download PDF
8. Genomic characterization of immune targets in patient-derived xenograft models for translational assessment of immunotherapy
- Author
-
Cerna, D., primary, Ciznadija, D., additional, Ben zvi, I., additional, Sloma, I., additional, Sidransky, D., additional, Davies, A., additional, and Goodwin, N., additional
- Published
- 2016
- Full Text
- View/download PDF
9. 334 - Genomic characterization of immune targets in patient-derived xenograft models for translational assessment of immunotherapy
- Author
-
Cerna, D., Ciznadija, D., Ben zvi, I., Sloma, I., Sidransky, D., Davies, A., and Goodwin, N.
- Published
- 2016
- Full Text
- View/download PDF
10. Ex vivo expansion of normal and chronic myeloid leukemic stem cells without functional alteration using a NUP98HOXA10homeodomain fusion gene
- Author
-
Sloma, I, primary, Imren, S, additional, Beer, P A, additional, Zhao, Y, additional, Lecault, V, additional, Leung, D, additional, Raghuram, K, additional, Brimacombe, C, additional, Lambie, K, additional, Piret, J, additional, Hansen, C, additional, Humphries, R K, additional, and Eaves, C J, additional
- Published
- 2012
- Full Text
- View/download PDF
11. Detection of clinically relevant variants in the TP53 gene below 10% allelic frequency: A multicenter study by ERIC, the European Research Initiative on CLL.
- Author
-
Pavlova S, Malcikova J, Radova L, Bonfiglio S, Cowland JB, Brieghel C, Andersen MK, Karypidou M, Biderman B, Doubek M, Lazarian G, Rapado I, Vynck M, Porret NA, Andres M, Rosenberg D, Sahar D, Martínez-Laperche C, Buño I, Hindley A, Donaldson D, Sánchez JB, García-Marco JA, Serrano-Alcalá A, Ferrer-Lores B, Fernández-Rodriguez C, Bellosillo B, Stilgenbauer S, Tausch E, Nikdin H, Quinn F, Atkinson E, van de Corput L, Yildiz C, Bilbao-Sieyro C, Florido Y, Thiede C, Schuster C, Stoj A, Czekalska S, Chatzidimitriou A, Laidou S, Bidet A, Dussiau C, Nollet F, Piras G, Monne M, Smirnova S, Nikitin E, Sloma I, Claudel A, Largeaud L, Ysebaert L, Valk PJM, Christian A, Walewska R, Oscier D, Sebastião M, da Silva MG, Galieni P, Angelini M, Rossi D, Spina V, Matos S, Martins V, Stokłosa T, Pepek M, Baliakas P, Andreu R, Luna I, Kahre T, Murumets Ü, Pikousova T, Kurucova T, Laird S, Ward D, Alcoceba M, Balanzategui A, Scarfo L, Gandini F, Zapparoli E, Blanco A, Abrisqueta P, Rodríguez-Vicente AE, Benito R, Bravetti C, Davi F, Gameiro P, Martinez-Lopez J, Tazón-Vega B, Baran-Marszak F, Davis Z, Catherwood M, Sudarikov A, Rosenquist R, Niemann CU, Stamatopoulos K, Ghia P, and Pospisilova S
- Abstract
In chronic lymphocytic leukemia, the reliability of next-generation sequencing (NGS) to detect TP53 variants ≤10% allelic frequency (low-VAF) is debated. We tested the ability to detect 23 such variants in 41 different laboratories using their NGS method of choice. The sensitivity was 85.6%, 94.5%, and 94.8% at 1%, 2%, and 3% VAF cut-off, respectively. While only one false positive (FP) result was reported at >2% VAF, it was more challenging to distinguish true variants <2% VAF from background noise (37 FPs reported by 9 laboratories). The impact of low-VAF variants on time-to-second-treatment (TTST) and overall survival (OS) was investigated in a series of 1092 patients. Among patients not treated with targeted agents, patients with low-VAF TP53 variants had shorter TTST and OS versus wt- TP53 patients, and the relative risk of second-line treatment or death increased continuously with increasing VAF. Targeted therapy in ≥2 line diminished the difference in OS between patients with low-VAF TP53 variants and wt- TP53 patients, while patients with high-VAF TP53 variants had inferior OS compared to wild type- TP53 cases. Altogether, NGS-based approaches are technically capable of detecting low-VAF variants. No strict threshold can be suggested from a technical standpoint, laboratories reporting TP53 mutations should participate in a standardized validation set-up. Finally, whereas low-VAF variants affected outcomes in patients receiving chemoimmunotherapy, their impact on those treated with novel therapies remains undetermined. Our results pave the way for the harmonized and accurate TP53 assessment, which is indispensable for elucidating the role of TP53 mutations in targeted treatment., Competing Interests: Bárbara Tazón‐Vega: Honoraria: Bristol Meyer Squibb. Beatriz Bellosillo: Advisory board honoraria, research support, travel support, speaker fees: Astra‐Zeneca, BMS, Janssen, Merck‐Serono, Novartis, Pfizer, Hoffman‐La Roche, ThermoFisher. Christian Brieghel: Travel grant: Octapharma. Carsten U. Niemann: Research funding and/or consultancy fees: Abbvie, AstraZeneca, Beigene, Janssen, Genmab, Lilly, MSD, CSL Behring, Takeda, Octapharma. Davide Rossi: Honoraria: AbbVie, AstraZeneca, BeiGene, BMS, Janssen, Lilly. Research grants: AbbVie, AstraZeneca, Janssen. Travel grants: AstraZeneca, Janssen. Eugen Tausch: Honoraria and research support: Abbvie, AstraZeneca, BeiGene, Janssen, Hoffmann‐La Roche; Research support from Abbvie, Roche, Gilead. Frédéric Davi: Honoraria: Janssen, AstraZeneca. Kostas Stamatopoulos: Research funding, honoraria and/or consultancy fees: Abbvie, AstraZeneca, Janssen, Lilly, Roche. Lydia Scarfo: Consultancy: AbbVie, AstraZeneca, BeiGene, Janssen, Lilly; Speaker Bureau: Octapharma. Miguel Alcoceba: Honoraria and travel grants: Janssen, AstraZeneca. Martin Andres: Consultancy, Honoraria, and travel support: AstraZeneca, Novartis, Roche, Janssen‐Cilag. Maria Gomes da Silva: Consultancy and Research Funding: Janssen Cilag, AstaZeneca, Abbvie, Roche, Takeda. Pau Abrisqueta: Consultancy and Honoraria: Janssen, Abbvie, Roche, BMS, AstraZeneca, Genmab. Panagiotis Baliakas: Honoraria: Abbvie, Gilead, Janssen. Research funding: Gilead. Paolo Ghia: Honoraria: AbbVie, Astrazeneca, BeiGene, BMS, Galapagos, Janssen, Lilly/Loxo Oncology, MSD, Roche. Research funding: AbbVie, AstraZeneca, BMS, Janssen. Richard Rosenquist: Honoraria: AbbVie, AstraZeneca, Janssen, Illumina, Roche. Renata Walewska: Travel support: AbbVie, AstraZeneca, Janssen, Beigene. Sylwia Czekalska: Honoraria: AstraZeneca. Funding: Janssen, AstraZeneca. Stephan Stilgenbauer: Advisory board honoraria, research support, travel support, speaker fees: AbbVie, Acerta, Amgen, AstraZeneca, BeiGene, BMS, Celgene, Gilead, GSK, Hoffmann‐La Roche, Infinity, Janssen, Lilly, Novartis, Sunesis, Verastem. Tiina Kahre: Honoraria: AstraZeneca. Tomasz Stokłosa: Honoraria and Research Funding: Janssen, AstraZeneca. The remaining authors have no competing interests to declare., (© 2025 The Author(s). HemaSphere published by John Wiley & Sons Ltd on behalf of European Hematology Association.)
- Published
- 2025
- Full Text
- View/download PDF
12. Mutation of Epigenetic Regulators at Diagnosis Is an Independent Predictor of Tyrosine Kinase Inhibitor Treatment Failure in Chronic Myeloid Leukemia: A Report From the RESIDIAG Study.
- Author
-
Guerineau H, Cayuela JM, Dulucq S, Tran Quang V, Tarfi S, Gricourt G, Barathon Q, Joy C, Wagner-Ballon O, Morisset S, Nicolini FE, Brunet E, Maury S, Roy L, Etienne G, Réa D, and Sloma I
- Published
- 2024
- Full Text
- View/download PDF
13. YAP1 induces bladder cancer progression and promotes immune evasion through IL-6/STAT3 pathway and CXCL deregulation.
- Author
-
Sadhukhan P, Feng M, Illingworth E, Sloma I, Ooki A, Matoso A, Sidransky D, Johnson BA 3rd, Marchionni L, Sillé FC, Choi W, McConkey D, and Hoque M
- Subjects
- Humans, Animals, Mice, Cell Line, Tumor, Tumor Microenvironment immunology, Transcription Factors metabolism, Transcription Factors genetics, Transcription Factors immunology, Tumor Escape, Neoplasm Proteins immunology, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Gene Expression Regulation, Neoplastic, Urinary Bladder Neoplasms immunology, Urinary Bladder Neoplasms pathology, Urinary Bladder Neoplasms genetics, Urinary Bladder Neoplasms metabolism, YAP-Signaling Proteins genetics, STAT3 Transcription Factor metabolism, STAT3 Transcription Factor genetics, STAT3 Transcription Factor immunology, Interleukin-6 metabolism, Interleukin-6 genetics, Interleukin-6 immunology, Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Adaptor Proteins, Signal Transducing immunology, Signal Transduction immunology
- Abstract
The Hippo signaling pathway plays a key role in tumorigenesis in different cancer types. We investigated the role of the Hippo effector YAP1 in the tumor immune microenvironment (TIME) of urothelial carcinoma of the bladder (UCB) and evaluated the efficacy of immunotherapy in the context of YAP1 signaling. We performed numerous in vitro and in vivo experiments to determine the role of YAP1 using genetic and pharmacological attenuation of YAP1 activity. Briefly, RNA sequencing was carried out with mouse and human cell lines to identify novel YAP1-regulated downstream targets unbiasedly. We then experimentally confirmed that YAP1 regulates the TIME through the IL-6/STAT3 signaling pathway and varied C-X-C motif chemokine regulation. We analyzed several human sample sets to explore the TIME status in the context of YAP1 expression. Our data indicate that YAP1 attenuation decreases M2 macrophages and myeloid-derived suppressor cells in the TIME compared with YAP1-expressing cells. In summary, this study provides insights into YAP1 signaling as a driver for cancer stemness and an inducer of immunosuppressive TIME. Moreover, the therapeutic efficacy of YAP1 attenuation indicates that combined blockade of YAP1 and immune checkpoints may yield clinical value for treating patients with UCB.
- Published
- 2024
- Full Text
- View/download PDF
14. Predicting which subsets of patients with myelodysplastic neoplasms are more likely to progress to overt chronic myelomonocytic leukemia.
- Author
-
Tran Quang V, Wagner-Ballon O, and Sloma I
- Subjects
- Humans, Prognosis, Biomarkers, Tumor genetics, Mutation, Dioxygenases, DNA-Binding Proteins, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic diagnosis, Leukemia, Myelomonocytic, Chronic pathology, Myelodysplastic Syndromes diagnosis, Myelodysplastic Syndromes genetics, Myelodysplastic Syndromes pathology, Disease Progression
- Abstract
The boundary between myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML) has been revised in the latest World Health Organization classification of myeloid malignancies. These changes were motivated by the description of a subgroup of MDS patients identified as oligomonocytic chronic myelomonocytic leukemia (OM-CMML) at risk of evolving into overt CMML. Various studies will be reviewed describing the clinical and biological features of MDS patients evolving to CMML. The efforts to discover biomarkers enabling the identification of these patients at the time of MDS diagnosis will be discussed. Finally, the molecular landscape of these patients will be presented with a specific focus on the biallelic inactivation of TET2 in light of its functional impact on hematopoietic stem cells, granule-monocytic differentiation, and its tight interplay with inflammation.
- Published
- 2024
- Full Text
- View/download PDF
15. Pr Connie J. Eaves (1944–2024).
- Author
-
Audet J, Berger M, Cashman J, Chalandon Y, Coulombel L, Flamant S, Ghaffari S, Lefort S, Lemoine F, Maguer-Satta V, Nicolini FE, Schmitt C, Sloma I, and Turhan A
- Published
- 2024
- Full Text
- View/download PDF
16. Long-term outcome after autologous BCR::ABL1 -negative peripheral blood stem cell transplantation in adults with Philadelphia-positive acute lymphoblastic leukemia: a comparative study.
- Author
-
Caillot L, Leclerc M, Sleiman EJR, Sloma I, Wagner-Ballon O, Claudel A, Beckerich F, Redjoul R, Robin C, Parinet V, Pautas C, Menouche D, Bouledroua S, Cabanne L, Nait-Sidenas Y, Gautier E, Rouard H, Lafon I, Chalandon Y, Boissel N, Caillot D, and Maury S
- Subjects
- Adult, Humans, Fusion Proteins, bcr-abl genetics, Philadelphia Chromosome, Peripheral Blood Stem Cell Transplantation, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy
- Published
- 2024
- Full Text
- View/download PDF
17. TET2 mutational status affects myelodysplastic syndrome evolution to chronic myelomonocytic leukemia.
- Author
-
Quang VT, Podvin B, Desterke C, Tarfi S, Barathon Q, Badaoui B, Freynet N, Parinet V, Leclerc M, Maury S, Solary E, Selimoglu-Buet D, Duployez N, Wagner-Ballon O, and Sloma I
- Subjects
- Humans, Mutation, DNA-Binding Proteins genetics, Leukemia, Myelomonocytic, Chronic genetics, Myelodysplastic Syndromes genetics, Dioxygenases
- Published
- 2023
- Full Text
- View/download PDF
18. ABO blood barrier to engraftment after allogeneic stem cell transplantation in sickle cell disease: A case-story with two successive HLA-matched sibling donors.
- Author
-
Redjoul R, Beckerich F, de Luna G, Leclerc M, Hebert N, Sloma I, Menouche D, Bartolucci P, and Maury S
- Subjects
- Humans, Siblings, Transplantation Conditioning, Hematopoietic Stem Cell Transplantation, Anemia, Sickle Cell therapy, Graft vs Host Disease
- Published
- 2023
- Full Text
- View/download PDF
19. Effective use of intensive treatment against multiple myeloma of recipient origin after allogeneic transplantation for acute myeloid leukemia.
- Author
-
de Charry F, Konopacki J, Bugier S, Foissaud V, Sloma I, Malfuson JV, and Arnautou P
- Abstract
We describe here a 56-years -old woman cured in our institution for an acute myeloid leukemia (AML) and a monoclonal gammopathy of undetermined significance (MGUS). In order to treat AML, underwent allogeneic stem cell transplantation in second complete remission. Four years after transplant, MGUS evolved to multiple myeloma and was intensively treated with "autologous" transplant after successful mobilization. This report illustrates: (i) a lack of efficacy of graft versus myeloma effect in a patient probably cured of AML by graft versus leukaemia effect; (ii) the ability to mobilize peripheral blood stem cells in order to perform "autologous" transplantation after allogeneic transplantation., Competing Interests: The authors have no financial disclosure or funding and no conflict of interest., (© 2023 The Authors. Published by Elsevier Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
20. Evaluation of two new highly multiplexed PCR assays as an alternative to next-generation sequencing for IDH1/2 mutation detection.
- Author
-
Favre L, Sako N, Tarfi S, Quang VT, Joy C, Dupuy A, Guillerm E, Gaulard P, Wagner-Ballon O, Pujals A, and Sloma I
- Subjects
- Humans, Mutation genetics, Polymerase Chain Reaction methods, Gene Frequency, Isocitrate Dehydrogenase genetics, High-Throughput Nucleotide Sequencing
- Abstract
IDH1 and IDH2 somatic mutations have been identified in solid tumors and blood malignancies. The development of inhibitors of mutant IDH1 and IDH2 in the past few years has prompted the development of a fast and sensitive assay to detect IDH1
R132 , IDH2R140 and IDH2R172 mutations to identify patients eligible for these targeted therapies. This study aimed to compare two new multiplexed PCR assays - an automated quantitative PCR (qPCR) on the PGX platform and a droplet digital PCR (ddPCR) with next-generation sequencing (NGS) for IDH1/2 mutation detection. These assays were evaluated on 102 DNA extracted from patient peripheral blood, bone marrow and formalin-fixed paraffin-embedded tissue samples with mutation allelic frequency ranging from 0.6% to 45.6%. The ddPCR assay had better analytical performances than the PGX assay with 100% specificity, 100% sensitivity and a detection limit down to 0.5% on IDH1R132 , IDH2R140 and IDH2R172 codons, and a high correlation with NGS results. Therefore, the new highly multiplexed ddPCR is a fast and cost-effective assay that meets most clinical needs to identify and follow cancer patients in the era of anti-IDH1/2-targeted therapies., (© 2022 The Authors. Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)- Published
- 2022
- Full Text
- View/download PDF
21. Onset of blast crisis in chronic myeloid leukemia patients in treatment-free remission.
- Author
-
Dulucq S, Hayette S, Cayuela JM, Bauduer F, Chabane K, Chevallier P, Cony-Makhoul P, Flandrin-Gresta P, Jeune CL, Bris YL, Legros L, Maisonneuve H, Roy L, Mahon FX, Sloma I, Rea D, and Nicolini FE
- Subjects
- Humans, Imatinib Mesylate, Remission Induction, Blast Crisis, Leukemia, Myelogenous, Chronic, BCR-ABL Positive diagnosis, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy
- Published
- 2022
- Full Text
- View/download PDF
22. [Myeloid neoplasms associated with rearrangement of PDGFRB: A rare and tricky disease].
- Author
-
Bontoux C, Badaoui B, Abermil N, Tarfi S, Guermouche H, Dubois S, Roy L, Xuan JV, Quang VT, Wang L, Favre L, Poullot E, Michel M, Sloma I, Crickx E, and Pécriaux A
- Subjects
- Humans, Male, Aged, Receptor, Platelet-Derived Growth Factor beta genetics, Imatinib Mesylate therapeutic use, In Situ Hybridization, Fluorescence, Immunoglobulins, Intravenous genetics, Myeloproliferative Disorders complications, Myeloproliferative Disorders diagnosis, Myeloproliferative Disorders genetics, Eosinophilia genetics, Eosinophilia diagnosis, Eosinophilia therapy, Hematologic Neoplasms
- Abstract
In the latest World Health Organization classification (WHO), eosinophilic disorders represent a group of rare pathologic conditions with highly heterogeneous pathophysiology. In this report, we describe a case of myeloid neoplasm associated with eosinophilia and rearrangement of PDGFRB gene in a 67-year-old-male patient hospitalized with cerebellous ataxia. Initial investigations showed a bicytopenia with hypereosinophilia varying from 1.1 to 1.6×10
9 /L. Bone marrow aspiration was rich and showed a heterogeneous distribution of myeloid cells with clusters of promyelocytes and proerythroblasts associated with numerous eosinophils and spindle-shaped mast cells but without excess of blasts, dysplasia nor maturation skewing. These aspects suggested an atypical myeloproliferative neoplasm. Bone marrow biopsy was performed showing also a very high cellularity with area of myeloid and erythroid precursors associated with numerous spindle-shaped mast cells. Diagnoses of unclassified myeloid neoplasm and/or systemic mastocytosis were then proposed. Further chromosome analysis showed a t(5;8) translocation with PDGFRB rearrangement revealed in fluorescent in situ hybridization. Patient was treated with imatinib and intravenous immunoglobulin therapy allowing a significant improvement in neurological symptoms and biological results. Patient condition is currently stable after six lines of treatment. This rare hematopoietic neoplasm displays unusual histological and cytological features and can mimic other myeloproliferative neoplasm. Specific cytogenetics analysis should be considered for such cases with hypereosinophilia to select patients that may benefit from targeted therapy., (Copyright © 2022 Elsevier Masson SAS. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
23. Fusion Gene Detection and Quantification by Asymmetric Capture Sequencing (aCAP-Seq).
- Author
-
Gricourt G, Tran Quang V, Cayuela JM, Boudali E, Tarfi S, Barathon Q, Daveau R, Joy C, Wagner-Ballon O, Bories D, Pautas C, Maury S, Rea D, Roy L, and Sloma I
- Subjects
- Humans, High-Throughput Nucleotide Sequencing methods, Mutation genetics, Recurrence, Fusion Proteins, bcr-abl genetics, Leukemia, Myelogenous, Chronic, BCR-ABL Positive diagnosis, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy
- Abstract
Several fusion genes such as BCR::ABL1, FIP1L1::PDGFRA, and PML::RARA are now efficiently targeted by specific therapies in patients with leukemia. Although these therapies have significantly improved patient outcomes, leukemia relapse and progression remain clinical concerns. Most myeloid next-generation sequencing (NGS) panels do not detect or quantify these fusions. It therefore remains difficult to decipher the clonal architecture and dynamics of myeloid malignancy patients, although these factors can affect clinical decisions and provide pathophysiologic insights. An asymmetric capture sequencing strategy (aCAP-Seq) and a bioinformatics algorithm (HmnFusion) were developed to detect and quantify MBCR::ABL1, μBCR::ABL1, PML::RARA, and FIP1L1::PDGFRA fusion genes in an NGS panel targeting 41 genes. One-hundred nineteen DNA samples derived from 106 patients were analyzed by conventional methods at diagnosis or on follow-up and were sequenced with this NGS myeloid panel. The specificity and sensitivity of fusion detection by aCAP-Seq were 100% and 98.1%, respectively, with a limit of detection estimated at 0.1%. Fusion quantifications were linear from 0.1% to 50%. Breakpoint locations and sequences identified by NGS were concordant with results obtained by Sanger sequencing. Finally, this new sensitive and cost-efficient NGS method allowed integrated analysis of resistant chronic myeloid leukemia patients and thus will be of interest to elucidate the mutational landscape and clonal architecture of myeloid malignancies driven by these fusion genes at diagnosis, relapse, or progression., (Copyright © 2022 Association for Molecular Pathology and American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
24. High prevalence of unusual KRAS, NRAS, and BRAF mutations in POLE-hypermutated colorectal cancers.
- Author
-
Favre L, Cohen J, Calderaro J, Pécriaux A, Nguyen CT, Bourgoin R, Larnaudie L, Dupuy A, Ollier M, Lechapt E, Sloma I, Tournigand C, Rousseau B, and Pujals A
- Subjects
- Biomarkers, Tumor genetics, GTP Phosphohydrolases genetics, Humans, Male, Membrane Proteins genetics, Mutation genetics, Prevalence, Proto-Oncogene Proteins p21(ras) genetics, Colorectal Neoplasms diagnosis, Colorectal Neoplasms epidemiology, Colorectal Neoplasms genetics, DNA Polymerase II genetics, Poly-ADP-Ribose Binding Proteins genetics, Proto-Oncogene Proteins B-raf genetics
- Abstract
Exonucleasic domain POLE (edPOLE) mutations, which are responsible for a hypermutated tumor phenotype, occur in 1-2% of colorectal cancer (CRC) cases. These alterations represent an emerging biomarker for response to immune checkpoint blockade. This study aimed to assess the molecular characteristics of edPOLE-mutated tumors to facilitate patient screening. Based on opensource data analysis, we compared the prevalence of edPOLE mutations in a control group of unselected CRC patients (n = 222) vs a group enriched for unusual BRAF/RAS mutations (n = 198). Tumor mutational burden (TMB) and immune infiltrate of tumors harboring edPOLE mutations were then analyzed. In total, 420 CRC patients were analyzed: 11 edPOLE-mutated tumors were identified, most frequently in microsatellite (MMR)-proficient young (< 70 years) male patients, with left-sided tumors harboring noncodon 12 KRAS mutation. The prevalence of edPOLE-mutated tumors in the control vs the experimental screening group was, respectively, 0.45% (n = 1) vs 5.0% (n = 10). Among the 11 edPOLE-mutated cases, two had a low TMB, three were hypermutated, and six were ultramutated. EdPOLE-mutated cases had a high CD8
+ tumor-infiltrating lymphocyte (TIL) infiltration. These clinicopathological and molecular criteria may help to identify edPOLE mutations associated with a high TMB in CRC, and improve the selection of patients who could benefit from immunotherapy., (© 2022 The Authors. Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)- Published
- 2022
- Full Text
- View/download PDF
25. Acquired spherocytosis due to somatic ANK1 mutations as a manifestation of clonal hematopoiesis in elderly patients.
- Author
-
Mansour-Hendili L, Flamarion E, Michel M, Morbieu C, Gameiro C, Sloma I, Badaoui B, Darnige L, Camard M, Lunati-Rozie A, Aissat A, Tarfi S, Friedrich C, Picard V, Garçon L, Abermil N, Kaltenbach S, Radford-Weiss I, Kosmider O, Fanen P, Bartolucci P, Godeau B, Galactéros F, and Funalot B
- Subjects
- Aged, Ankyrins genetics, Hematopoiesis, Humans, Clonal Hematopoiesis, Mutation, Spherocytosis, Hereditary genetics
- Published
- 2022
- Full Text
- View/download PDF
26. Leukaemic evolution of rare donor CSF3R germline mutation after umbilical cord blood transplant.
- Author
-
Arnautou P, Quang VT, Konopacki J, Chevillon F, Bories D, Sloma I, and Malfuson JV
- Subjects
- Fetal Blood, Germ-Line Mutation, Humans, Receptors, Colony-Stimulating Factor, Signal Transduction, Cord Blood Stem Cell Transplantation
- Published
- 2022
- Full Text
- View/download PDF
27. Functional Genomic Identification of Predictors of Sensitivity and Mechanisms of Resistance to Multivalent Second-Generation TRAIL-R2 Agonists.
- Author
-
Grinkevitch V, Wappett M, Crawford N, Price S, Lees A, McCann C, McAllister K, Prehn J, Young J, Bateson J, Gallagher L, Michaut M, Iyer V, Chatzipli A, Barthorpe S, Ciznadija D, Sloma I, Wesa A, Tice DA, Wessels L, Garnett M, Longley DB, McDermott U, and McDade SS
- Subjects
- Apoptosis, CASP8 and FADD-Like Apoptosis Regulating Protein genetics, CASP8 and FADD-Like Apoptosis Regulating Protein metabolism, Caspase 8 genetics, Cell Line, Tumor, Genomics, Humans, Proto-Oncogene Proteins c-bcl-2 metabolism, TNF-Related Apoptosis-Inducing Ligand metabolism, TNF-Related Apoptosis-Inducing Ligand pharmacology
- Abstract
Multivalent second-generation TRAIL-R2 agonists are currently in late preclinical development and early clinical trials. Herein, we use a representative second-generation agent, MEDI3039, to address two major clinical challenges facing these agents: lack of predictive biomarkers to enable patient selection and emergence of resistance. Genome-wide CRISPR knockout screens were notable for the lack of resistance mechanisms beyond the canonical TRAIL-R2 pathway (caspase-8, FADD, BID) as well as p53 and BAX in TP53 wild-type models, whereas a CRISPR activatory screen identified cell death inhibitors MCL-1 and BCL-XL as mechanisms to suppress MEDI3039-induced cell death. High-throughput drug screening failed to identify genomic alterations associated with response to MEDI3039; however, transcriptomics analysis revealed striking association between MEDI3039 sensitivity and expression of core components of the extrinsic apoptotic pathway, most notably its main apoptotic effector caspase-8 in solid tumor cell lines. Further analyses of colorectal cell lines and patient-derived xenografts identified caspase-8 expression ratio to its endogenous regulator FLIP(L) as predictive of sensitivity to MEDI3039 in several major solid tumor types and a further subset indicated by caspase-8:MCL-1 ratio. Subsequent MEDI3039 combination screening of TRAIL-R2, caspase-8, FADD, and BID knockout models with 60 compounds with varying mechanisms of action identified two inhibitor of apoptosis proteins (IAP) that exhibited strong synergy with MEDI3039 that could reverse resistance only in BID-deleted models. In summary, we identify the ratios of caspase-8:FLIP(L) and caspase-8:MCL-1 as potential predictive biomarkers for second-generation TRAIL-R2 agonists and loss of key effectors such as FADD and caspase-8 as likely drivers of clinical resistance in solid tumors., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
28. Functional impact of a germline RET mutation in alveolar rhabdomyosarcoma.
- Author
-
Berlow NE, Crawford KA, Bult CJ, Noakes C, Sloma I, Rudzinski ER, and Keller C
- Subjects
- Animals, Cell Line, Tumor, DNA Mutational Analysis, Genotype, Humans, Mice, Multiple Endocrine Neoplasia Type 2a genetics, Multiple Endocrine Neoplasia Type 2a pathology, Phenotype, Rhabdomyosarcoma, Alveolar drug therapy, Xenograft Model Antitumor Assays, Germ Cells physiology, Germ-Line Mutation, Proto-Oncogene Proteins c-ret genetics, Proto-Oncogene Proteins c-ret metabolism, Rhabdomyosarcoma, Alveolar genetics
- Abstract
Specific mutations in the RET proto-oncogene are associated with multiple endocrine neoplasia type 2A, a hereditary syndrome characterized by tumorigenesis in multiple glandular elements. In rare instances, MEN2A-associated germline RET mutations have also occurred with non-MEN2A associated cancers. One such germline mutant RET mutation occurred concomitantly in a young adult diagnosed with alveolar rhabdomyosarcoma, a pediatric and young adult soft-tissue cancer with a generally poor prognosis. Although tumor tissue samples were initially unable to provide a viable cell culture for study, tumor tissues were sequenced for molecular characteristics. Through a hierarchical clustering approach, the index case sample was matched to several genetically similar cell models, which were transformed to express the same mutant RET as the index case and used to explore potential therapeutic options for mutant RET -bearing alveolar rhabdomyosarcoma. We also determined whether the RET mutation associated with the index case caused synthetic lethality to select clinical agents. From our investigation, we did not identify synthetic lethality associated with the expression of that patient's RET variant, and overall we did not find experimental evidence for the role of RET in rhabdomyosarcoma progression., (© 2021 Berlow et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2021
- Full Text
- View/download PDF
29. Genomic analysis of primary and secondary myelofibrosis redefines the prognostic impact of ASXL1 mutations: a FIM study.
- Author
-
Luque Paz D, Riou J, Verger E, Cassinat B, Chauveau A, Ianotto JC, Dupriez B, Boyer F, Renard M, Mansier O, Murati A, Rey J, Etienne G, Mansat-De Mas V, Tavitian S, Nibourel O, Girault S, Le Bris Y, Girodon F, Ranta D, Chomel JC, Cony-Makhoul P, Sujobert P, Robles M, Ben Abdelali R, Kosmider O, Cottin L, Roy L, Sloma I, Vacheret F, Wemeau M, Mossuz P, Slama B, Cussac V, Denis G, Walter-Petrich A, Burroni B, Jézéquel N, Giraudier S, Lippert E, Socié G, Kiladjian JJ, and Ugo V
- Subjects
- Bayes Theorem, Genomics, Humans, Mutation, Prognosis, Repressor Proteins genetics, Primary Myelofibrosis diagnosis, Primary Myelofibrosis genetics
- Abstract
We aimed to study the prognostic impact of the mutational landscape in primary and secondary myelofibrosis. The study included 479 patients with myelofibrosis recruited from 24 French Intergroup of Myeloproliferative Neoplasms (FIM) centers. The molecular landscape was studied by high-throughput sequencing of 77 genes. A Bayesian network allowed the identification of genomic groups whose prognostic impact was studied in a multistate model considering transitions from the 3 conditions: myelofibrosis, acute leukemia, and death. Results were validated using an independent, previously published cohort (n = 276). Four genomic groups were identified: patients with TP53 mutation; patients with ≥1 mutation in EZH2, CBL, U2AF1, SRSF2, IDH1, IDH2, NRAS, or KRAS (high-risk group); patients with ASXL1-only mutation (ie, no associated mutation in TP53 or high-risk genes); and other patients. A multistate model found that both TP53 and high-risk groups were associated with leukemic transformation (hazard ratios [HRs] [95% confidence interval], 8.68 [3.32-22.73] and 3.24 [1.58-6.64], respectively) and death from myelofibrosis (HRs, 3.03 [1.66-5.56] and 1.77 [1.18-2.67], respectively). ASXL1-only mutations had no prognostic value that was confirmed in the validation cohort. However, ASXL1 mutations conferred a worse prognosis when associated with a mutation in TP53 or high-risk genes. This study provides a new definition of adverse mutations in myelofibrosis with the addition of TP53, CBL, NRAS, KRAS, and U2AF1 to previously described genes. Furthermore, our results argue that ASXL1 mutations alone cannot be considered detrimental., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
30. Epigenetic and functional changes imposed by NUP98-HOXA9 in a genetically engineered model of chronic myeloid leukemia progression.
- Author
-
Sloma I, Beer P, Desterke C, Bulaeva E, Bilenky M, Carles A, Moksa M, Raghuram K, Brimacombe C, Lambie K, Turhan AG, Wagner-Ballon O, Gaulard P, Humphries K, Hirst M, and Eaves CJ
- Subjects
- Epigenesis, Genetic, Homeodomain Proteins genetics, Humans, Nuclear Pore Complex Proteins genetics, Oncogene Proteins, Fusion genetics, Translocation, Genetic, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Leukemia, Myeloid genetics
- Published
- 2021
- Full Text
- View/download PDF
31. Prognostic value of monocyte subset distribution in chronic myelomonocytic leukemia: results of a multicenter study.
- Author
-
Jestin M, Tarfi S, Duchmann M, Badaoui B, Freynet N, Tran Quang V, Sloma I, Droin N, Morabito M, Leclerc M, Maury S, Fenaux P, Solary E, Selimoglu-Buet D, and Wagner-Ballon O
- Subjects
- Humans, Leukemia, Myelomonocytic, Chronic classification, Leukemia, Myelomonocytic, Chronic metabolism, Prognosis, Survival Rate, Leukemia, Myelomonocytic, Chronic pathology, Monocytes pathology
- Published
- 2021
- Full Text
- View/download PDF
32. Cholesterol Pathway Inhibition Induces TGF-β Signaling to Promote Basal Differentiation in Pancreatic Cancer.
- Author
-
Gabitova-Cornell L, Surumbayeva A, Peri S, Franco-Barraza J, Restifo D, Weitz N, Ogier C, Goldman AR, Hartman TR, Francescone R, Tan Y, Nicolas E, Shah N, Handorf EA, Cai KQ, O'Reilly AM, Sloma I, Chiaverelli R, Moffitt RA, Khazak V, Fang CY, Golemis EA, Cukierman E, and Astsaturov I
- Subjects
- 3-Hydroxysteroid Dehydrogenases genetics, 3-Hydroxysteroid Dehydrogenases metabolism, Animals, Atorvastatin pharmacology, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal metabolism, Cell Differentiation drug effects, Cell Differentiation genetics, Cell Line, Tumor, Epithelial-Mesenchymal Transition drug effects, Epithelial-Mesenchymal Transition genetics, Gene Expression Profiling, Gene Expression Regulation, Neoplastic drug effects, Humans, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, Kaplan-Meier Estimate, Mice, Inbred C57BL, Mice, Knockout, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms metabolism, Signal Transduction genetics, Transforming Growth Factor beta metabolism, Xenograft Model Antitumor Assays methods, Biosynthetic Pathways genetics, Carcinoma, Pancreatic Ductal genetics, Cholesterol, LDL biosynthesis, Pancreatic Neoplasms genetics, Transforming Growth Factor beta genetics
- Abstract
Oncogenic transformation alters lipid metabolism to sustain tumor growth. We define a mechanism by which cholesterol metabolism controls the development and differentiation of pancreatic ductal adenocarcinoma (PDAC). Disruption of distal cholesterol biosynthesis by conditional inactivation of the rate-limiting enzyme Nsdhl or treatment with cholesterol-lowering statins switches glandular pancreatic carcinomas to a basal (mesenchymal) phenotype in mouse models driven by Kras
G12D expression and homozygous Trp53 loss. Consistently, PDACs in patients receiving statins show enhanced mesenchymal features. Mechanistically, statins and NSDHL loss induce SREBP1 activation, which promotes the expression of Tgfb1, enabling epithelial-mesenchymal transition. Evidence from patient samples in this study suggests that activation of transforming growth factor β signaling and epithelial-mesenchymal transition by cholesterol-lowering statins may promote the basal type of PDAC, conferring poor outcomes in patients., Competing Interests: Declaration of Interests I.A. served as a consultant for Caris Life Sciences, Inc., (Copyright © 2020 Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
33. Disappearance of slan-positive non-classical monocytes for diagnosis of chronic myelomonocytic leukemia with an associated inflammatory state
- Author
-
Tarfi S, Badaoui B, Freynet N, Morabito M, Lafosse J, Toma A, Etienne G, Micol JB, Sloma I, Fenaux P, Solary E, Selimoglu-Buet D, and Wagner-Ballon O
- Published
- 2020
- Full Text
- View/download PDF
34. Volasertib preclinical activity in high-risk hepatoblastoma.
- Author
-
Kats D, Ricker CA, Berlow NE, Noblet B, Nicolle D, Mevel K, Branchereau S, Judde JG, Stiverson CD, Stiverson CL, Svalina MN, Settelmeyer T, Matlock K, Lathara M, Mussini C, Geller JI, Noakes C, Sloma I, Bharathy N, Cairo S, and Keller C
- Abstract
Relapsed and metastatic hepatoblastoma represents an unmet clinical need with limited chemotherapy treatment options. In a chemical screen, we identified volasertib as an agent with in vitro activity, inhibiting hepatoblastoma cell growth while sparing normal hepatocytes. Volasertib targets PLK1 and prevents the progression of mitosis, resulting in eventual cell death. PLK1 is overexpressed in hepatoblastoma biopsies relative to normal liver tissue. As a potential therapeutic strategy, we tested the combination of volasertib and the relapse-related hepatoblastoma chemotherapeutic irinotecan. We found both in vitro and in vivo efficacy of this combination, which may merit further preclinical investigation and exploration for a clinical trial concept., Competing Interests: CONFLICTS OF INTEREST The authors have no conflicts of interest with respect to the drugs used in this study., (Copyright: Kats et al.)
- Published
- 2019
- Full Text
- View/download PDF
35. Alternative Splicing of RAD6B and Not RAD6A is Selectively Increased in Melanoma: Identification and Functional Characterization.
- Author
-
Gajan A, Martin CE, Kim S, Joshi M, Michelhaugh SK, Sloma I, Mittal S, Firestine S, and Shekhar MPV
- Subjects
- Adult, Aged, Aged, 80 and over, Alternative Splicing, Cell Line, DNA Repair, DNA Replication, Female, Humans, Male, Melanoma diagnosis, Melanoma metabolism, Middle Aged, Skin Neoplasms diagnosis, Skin Neoplasms metabolism, Transcription, Genetic, Transcriptome, Ubiquitin-Conjugating Enzymes metabolism, Exome Sequencing methods, Wnt Signaling Pathway, beta Catenin metabolism, Melanoma, Cutaneous Malignant, Melanoma genetics, Skin Neoplasms genetics, Ubiquitin-Conjugating Enzymes genetics
- Abstract
Rad6B, a principal component of the translesion synthesis pathway, and activator of canonical Wnt signaling, plays an essential role in cutaneous melanoma development and progression. As Rad6 is encoded by two genes, namely, UBE2A ( RAD 6A ) and UBE2B ( R AD 6B ), in humans, we compared their expressions in melanomas and normal melanocytes. While both genes are weakly expressed in normal melanocytes, Rad6B is more robustly expressed in melanoma lines and patient-derived metastatic melanomas than RAD6A. The characterization of RAD6B transcripts revealed coexpression of various splice variants representing truncated or modified functional versions of wild-type RAD6B in melanomas, but not in normal melanocytes. Notably, two RAD6B isoforms with intact catalytic domains, RAD6BΔexon4 and RAD6Bintron5ins, were identified. We confirmed that RAD6BΔexon4 and RAD6Bintron5ins variants are expressed as 14 and 15 kDa proteins, respectively, with functional in vivo ubiquitin conjugating activity. Whole exome sequence analysis of 30 patient-derived melanomas showed RAD6B variants coexpressed with wild-type RAD6B in all samples analyzed, and RAD6Bintron5ins variants were found in half the cases. These variants constitute the majority of the RAD6B transcriptome in contrast to RAD6A, which was predominantly wild-type. The expression of functional RAD6B variants only in melanomas reveals RAD6B's molecular heterogeneity and its association with melanoma pathogenesis., Competing Interests: The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.
- Published
- 2019
- Full Text
- View/download PDF
36. Immunological analysis of phase II glioblastoma dendritic cell vaccine (Audencel) trial: immune system characteristics influence outcome and Audencel up-regulates Th1-related immunovariables.
- Author
-
Erhart F, Buchroithner J, Reitermaier R, Fischhuber K, Klingenbrunner S, Sloma I, Hibsh D, Kozol R, Efroni S, Ricken G, Wöhrer A, Haberler C, Hainfellner J, Krumpl G, Felzmann T, Dohnal AM, Marosi C, and Visus C
- Subjects
- Antigens, CD metabolism, Boron Compounds metabolism, Brain Neoplasms blood, Brain Neoplasms immunology, CD8-Positive T-Lymphocytes drug effects, Female, Glioblastoma blood, Glioblastoma immunology, Humans, Kaplan-Meier Estimate, Killer Cells, Natural drug effects, Killer Cells, Natural metabolism, Longitudinal Studies, Male, Treatment Outcome, Up-Regulation, Brain Neoplasms therapy, CD8-Positive T-Lymphocytes physiology, Cancer Vaccines therapeutic use, Dendritic Cells physiology, Glioblastoma therapy
- Abstract
Audencel is a dendritic cell (DC)-based cellular cancer immunotherapy against glioblastoma multiforme (GBM). It is characterized by loading of DCs with autologous whole tumor lysate and in vitro maturation via "danger signals". The recent phase II "GBM-Vax" trial showed no clinical efficacy for Audencel as assessed with progression-free and overall survival in all patients. Here we present immunological research accompanying the trial with a focus on immune system factors related to outcome and Audencel's effect on the immune system. Methodologically, peripheral blood samples (from apheresis before Audencel or venipuncture during Audencel) were subjected to functional characterization via enzyme-linked immunospot (ELISPOT) assays connected with cytokine bead assays (CBAs) as well as phenotypical characterization via flow cytometry and mRNA quantification. GBM tissue samples (from surgery) were subjected to T cell receptor sequencing and immunohistochemistry. As results we found: Patients with favorable pre-existing anti-tumor characteristics lived longer under Audencel than Audencel patients without them. Pre-vaccination blood CD8+ T cell count and ELISPOT Granzyme B production capacity in vitro upon tumor antigen exposure were significantly correlated with overall survival. Despite Audencel's general failure to induce a significant clinical response, it nevertheless seemed to have an effect on the immune system. For instance, Audencel led to a significant up-regulation of the Th1-related immunovariables ELISPOT IFNγ, the transcription factor T-bet in the blood and ELISPOT IL-2 in a dose-dependent manner upon vaccination. Post-vaccination levels of ELISPOT IFNγ and CD8+ cells in the blood were indicative of a significantly better survival. In summary, Audencel failed to reach an improvement of survival in the recent phase II clinical trial. No clinical efficacy was registered. Our concomitant immunological work presented here indicates that outcome under Audencel was influenced by the state of the immune system. On the other hand, Audencel also seemed to have stimulated the immune system. Overall, these immunological considerations suggest that DC immunotherapy against glioblastoma should be studied further - with the goal of translating an apparent immunological response into a clinical response. Future research should concentrate on investigating augmentation of immune reactions through combination therapies or on developing meaningful biomarkers.
- Published
- 2018
- Full Text
- View/download PDF
37. Aryl hydrocarbon receptor (AHR) is a novel druggable pathway controlling malignant progenitor proliferation in chronic myeloid leukemia (CML).
- Author
-
Gentil M, Hugues P, Desterke C, Telliam G, Sloma I, Souza LEB, Baykal S, Artus J, Griscelli F, Guerci A, Johnson-Ansah H, Foudi A, Bennaceur-Griscelli A, and Turhan AG
- Subjects
- Basic Helix-Loop-Helix Transcription Factors agonists, Basic Helix-Loop-Helix Transcription Factors genetics, Carbazoles pharmacology, Case-Control Studies, Cell Line, Tumor, Cell Proliferation drug effects, Fusion Proteins, bcr-abl genetics, Fusion Proteins, bcr-abl metabolism, Gene Expression Regulation, Neoplastic drug effects, Humans, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells pathology, Purines pharmacology, RNA, Messenger genetics, RNA, Messenger metabolism, RNA, Neoplasm genetics, RNA, Neoplasm metabolism, Receptors, Aryl Hydrocarbon agonists, Receptors, Aryl Hydrocarbon genetics, Signal Transduction drug effects, Tumor Stem Cell Assay, Basic Helix-Loop-Helix Transcription Factors metabolism, Leukemia, Myelogenous, Chronic, BCR-ABL Positive metabolism, Neoplastic Stem Cells metabolism, Receptors, Aryl Hydrocarbon metabolism
- Abstract
Aryl Hydrocarbon Receptor (AHR) is an ubiquitous basic helix-loop-helix transcription factor, which is ligand-activated and involved in numerous biological processes including cell division, cell quiescence and inflammation. It has been shown that AHR is involved in normal hematopoietic progenitor proliferation in human cells. In addition, loss of AHR in knockout mice is accompanied by a myeloproliferative syndrome-like disease, suggesting a role of AHR in hematopoietic stem cell (HSC) maintenance. To study the potential role of AHR pathway in CML progenitors and stem cells, we have first evaluated the expression of AHR in UT-7 cell line expressing BCR-ABL. AHR expression was highly reduced in UT-7 cell expressing BCR-ABL as compared to controls. AHR transcript levels, quantified in primary peripheral blood CML cells at diagnosis (n = 31 patients) were found to be significantly reduced compared to healthy controls (n = 15). The use of StemRegenin (SR1), an AHR antagonist, induced a marked expansion of total leukemic cells and leukemic CD34+ cells by about 4- and 10-fold respectively. SR1-treated CML CD34+ cells generated more colony-forming cells and long-term culture initiating cell (LTC-IC)-derived progenitors as compared to non-SR1-treated counterparts. Conversely, treatment of CML CD34+ cells with FICZ, a natural agonist of AHR, induced a 3-fold decrease in the number of CD34+ cells in culture after 7 days. Moreover, a 4-day FICZ treatment was sufficient to significantly reduce the clonogenic potential of CML CD34+ cells and this effect was synergized by Imatinib and Dasatinib treatments. Similarly, a 3-day FICZ treatment contributed to hinder significantly the number of LTC-IC-derived progenitors without synergistic effect with Imatinib. The analysis of molecular circuitry of AHR signaling in CML showed a transcriptional signature in CML derived CD34+ CD38- primitive cells with either low or high levels of AHR, with an upregulation of myeloid genes involved in differentiation in the "AHR low" fraction and an upregulation of genes involved in stem cell maintenance in the "AHR high" fraction. In conclusion, these findings demonstrate for the first time that down-regulation of AHR expression, a major cell cycle regulator, is involved in the myeloproliferative phenotype associated with CML. AHR agonists inhibit clonogenic and LTC-IC-derived progenitor growth and they could be used in leukemic stem cell targeting in CML., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2018
- Full Text
- View/download PDF
38. Whole-genome analysis reveals unexpected dynamics of mutant subclone development in a patient with JAK2-V617F-positive chronic myeloid leukemia.
- Author
-
Sloma I, Mitjavila-Garcia MT, Feraud O, Griscelli F, Oudrhiri N, El Marsafy S, Gobbo E, Divers D, Proust A, Smadja DM, Desterke C, Carles A, Ma Y, Hirst M, Marra MA, Eaves CJ, Bennaceur-Griscelli A, and Turhan AG
- Subjects
- Cell Cycle Proteins genetics, DNA-Binding Proteins genetics, Genome-Wide Association Study, Humans, Induced Pluripotent Stem Cells physiology, Male, Middle Aged, Nuclear Proteins genetics, Transcription Factors genetics, Janus Kinase 2 genetics, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Mutation
- Abstract
We report here the first use of whole-genome sequencing (WGS) to examine the initial clonal dynamics in an unusual patient with chronic myeloid leukemia (CML), who presented in chronic phase (CP) with doubly marked BCR-ABL1
+ /JAK2V617F -mutant cells and, over a 9-year period, progressed into an accelerated phase (AP) and then terminal blast phase (BP). WGS revealed that the diagnostic cells also contained mutations in ASXL1, SEC23B, MAD1L1, and RREB1 as well as 12,000 additional uncommon DNA variants. WGS of endothelial cells generated from circulating precursors revealed many of these were shared with the CML clone. Surprisingly, WGS of induced pluripotent stem cells (iPSCs) derived from the AP cells revealed only six additional coding somatic mutations, despite retention by the hematopoietic progeny of the parental AP cell levels of BCR-ABL1 expression and sensitivity to imatinib and pimozide. Limited analysis of BP cells revealed independent subclonal progression to homozygosity of the MAD1L1 and RREB1 variants. MAD1L1 and SEC23B mutations were also identified in 2 of 101 cases of myeloproliferative neoplasms, but not in 42 healthy subjects. These findings challenge historic concepts of clonal evolution in CML., (Copyright © 2017 ISEH - International Society for Experimental Hematology. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
39. In silico analysis of pathways activation landscape in oral squamous cell carcinoma and oral leukoplakia.
- Author
-
Makarev E, Schubert AD, Kanherkar RR, London N, Teka M, Ozerov I, Lezhnina K, Bedi A, Ravi R, Mehra R, Hoque MO, Sloma I, Gaykalova DA, Csoka AB, Sidransky D, Zhavoronkov A, and Izumchenko E
- Abstract
A subset of patients with oral squamous cell carcinoma (OSCC), the most common subtype of head and neck squamous cell carcinoma (HNSCC), harbor dysplastic lesions (often visually identified as leukoplakia) prior to cancer diagnosis. Although evidence suggest that leukoplakia represents an initial step in the progression to cancer, signaling networks driving this progression are poorly understood. Here, we applied in silico Pathway Activation Network Decomposition Analysis (iPANDA), a new bioinformatics software suite for qualitative analysis of intracellular signaling pathway activation using transcriptomic data, to assess a network of molecular signaling in OSCC and pre-neoplastic oral lesions. In tumor samples, our analysis detected major conserved mitogenic and survival signaling pathways strongly associated with HNSCC, suggesting that some of the pathways identified by our algorithm, but not yet validated as HNSCC related, may be attractive targets for future research. While pathways activation landscape in the majority of leukoplakias was different from that seen in OSCC, a subset of pre-neoplastic lesions has demonstrated some degree of similarity to the signaling profile seen in tumors, including dysregulation of the cancer-driving pathways related to survival and apoptosis. These results suggest that dysregulation of these signaling networks may be the driving force behind the early stages of OSCC tumorigenesis. While future studies with larger leukoplakia data sets are warranted to further estimate the values of this approach for capturing signaling features that characterize relevant lesions that actually progress to cancers, our platform proposes a promising new approach for detecting cancer-promoting pathways and tailoring the right therapy to prevent tumorigenesis., Competing Interests: EM, IO, KL, MT and AZ are affiliated with Insilico Medicine, Inc., a company engaged in aging research, which uses and provides both paid and free services using a variety of pathway activation scoring algorithms and hence may have competing financial interests. The remaining authors declare no conflict of interest.
- Published
- 2017
- Full Text
- View/download PDF
40. Comparative mutational landscape analysis of patient-derived tumour xenografts.
- Author
-
Brait M, Izumchenko E, Kagohara LT, Long S, Wysocki PT, Faherty B, Fertig EJ, Khor TO, Bruckheimer E, Baia G, Ciznadija D, Sloma I, Ben-Zvi I, Paz K, and Sidransky D
- Subjects
- Animals, High-Throughput Nucleotide Sequencing methods, Humans, Mice, Neoplasm Transplantation, Neoplasms pathology, Polymerase Chain Reaction methods, Reproducibility of Results, Tumor Cells, Cultured, DNA Mutational Analysis methods, Heterografts metabolism, Neoplasms genetics
- Abstract
Background: Screening of patients for cancer-driving mutations is now used for cancer prognosis, remission scoring and treatment selection. Although recently emerged targeted next-generation sequencing-based approaches offer promising diagnostic capabilities, there are still limitations. There is a pressing clinical need for a well-validated, rapid, cost-effective mutation profiling system in patient specimens. Given their speed and cost-effectiveness, quantitative PCR mutation detection techniques are well suited for the clinical environment. The qBiomarker mutation PCR array has high sensitivity and shorter turnaround times compared with other methods. However, a direct comparison with existing viable alternatives are required to assess its true potential and limitations., Methods: In this study, we evaluated a panel of 117 patient-derived tumour xenografts by the qBiomarker array and compared with other methods for mutation detection, including Ion AmpliSeq sequencing, whole-exome sequencing and droplet digital PCR., Results: Our broad analysis demonstrates that the qBiomarker's performance is on par with that of other labour-intensive and expensive methods of cancer mutation detection of frequently altered cancer-associated genes, and provides a foundation for supporting its consideration as an option for molecular diagnostics., Conclusions: This large-scale direct comparison and validation of currently available mutation detection approaches is extremely relevant for the current scenario of precision medicine and will lead to informed choice of screening methodologies, especially in lower budget conditions or time frame limitations.
- Published
- 2017
- Full Text
- View/download PDF
41. Drug reaction with eosinophilia and systemic symptoms (DRESS) induced by imatinib in chronic myeloid leukemia.
- Author
-
Vatel O, Aumont C, Mathy V, Petit M, Feriel J, Sloma I, Bennaceur-Griscelli A, and Turhan AG
- Subjects
- Antineoplastic Agents therapeutic use, Blood Cell Count, Drug Substitution, Fusion Proteins, bcr-abl genetics, Humans, Imatinib Mesylate therapeutic use, Leukemia, Myelogenous, Chronic, BCR-ABL Positive diagnosis, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Male, Middle Aged, Protein Kinase Inhibitors therapeutic use, Skin pathology, Treatment Outcome, Antineoplastic Agents adverse effects, Drug Hypersensitivity Syndrome diagnosis, Drug Hypersensitivity Syndrome etiology, Imatinib Mesylate adverse effects, Leukemia, Myelogenous, Chronic, BCR-ABL Positive complications, Protein Kinase Inhibitors adverse effects
- Published
- 2017
- Full Text
- View/download PDF
42. Leukemic stem cell persistence in chronic myeloid leukemia patients in deep molecular response induced by tyrosine kinase inhibitors and the impact of therapy discontinuation.
- Author
-
Chomel JC, Bonnet ML, Sorel N, Sloma I, Bennaceur-Griscelli A, Rea D, Legros L, Marfaing-Koka A, Bourhis JH, Ame S, Guerci-Bresler A, Rousselot P, and Turhan AG
- Subjects
- Adult, Aged, Aged, 80 and over, Dasatinib therapeutic use, Female, Humans, Imatinib Mesylate therapeutic use, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Male, Middle Aged, Neoplasm Recurrence, Local epidemiology, Neoplasm, Residual, Neoplastic Stem Cells drug effects, Protein Kinase Inhibitors therapeutic use, Remission Induction, Antineoplastic Agents therapeutic use, Leukemia, Myelogenous, Chronic, BCR-ABL Positive pathology, Neoplasm Recurrence, Local pathology, Neoplastic Stem Cells pathology
- Abstract
During the last decade, the use of tyrosine kinase inhibitor (TKI) therapy has modified the natural history of chronic myeloid leukemia (CML) allowing an increase of the overall and disease-free survival, especially in patients in whom molecular residual disease becomes undetectable. However, it has been demonstrated that BCR-ABL1- expressing leukemic stem cells (LSCs) persist in patients in deep molecular response. It has also been shown that the discontinuation of Imatinib leads to a molecular relapse in the majority of cases. To determine a possible relationship between these two phenomena, we have evaluated by clonogenic and long-term culture initiating cell (LTC-IC) assays, the presence of BCR-ABL1-expressing LSCs in marrow samples from 21 patients in deep molecular response for three years after TKI therapy (mean duration seven years). LSCs were detected in 4/21 patients. Discontinuation of TKI therapy in 13/21 patients led to a rapid molecular relapse in five patients (4 without detectable LSCs and one with detectable LSCs). No relapse occurred in the eight patients still on TKI therapy, whether LSCs were detectable or not. Thus, this study demonstrates for the first time the in vivo efficiency of TKIs, both in the progenitor and the LSC compartments. It also confirms the persistence of leukemic stem cells in patients in deep molecular response, certainly at the origin of relapses. Finally, it emphasizes the difficulty of detecting residual LSCs due to their rarity and their low BCR-ABL1 mRNA expression., Competing Interests: AGT: Research support from Novartis, Consultancy for Bristol Myers Squibb; PR: Research support from Bristol Myers Squibb and ARIAD Pharmaceuticals.
- Published
- 2016
- Full Text
- View/download PDF
43. Experimental Support for the Ecoimmunity Theory: Distinct Phenotypes of Nonlymphocytic Cells in SCID and Wild-Type Mice.
- Author
-
Ochayon DE, Baranovski BM, Malkin P, Schuster R, Kalay N, Ben-Hamo R, Sloma I, Levinson J, Brazg J, Efroni S, Lewis EC, and Nevo U
- Subjects
- Animals, Autoimmunity genetics, Histocompatibility Antigens Class I metabolism, Insulin metabolism, Interleukin-10 metabolism, Interleukin-1alpha metabolism, Interleukin-6 metabolism, Mice, Mice, SCID, Tumor Necrosis Factor-alpha metabolism, Autoimmunity physiology, Islets of Langerhans metabolism, Lymphocytes metabolism
- Abstract
Immune tolerance toward "self" is critical in multiple immune disorders. While there are several mechanisms to describe the involvement of immune cells in the process, the role of peripheral tissue cells in that context is not yet clear. The theory of ecoimmunity postulates that interactions between immune and tissue cells represent a predator-prey relationship. A lifelong interaction, shaped mainly during early ontogeny, leads to selection of nonimmune cell phenotypes. Normally, therefore, nonimmune cells that evolve alongside an intact immune system would be phenotypically capable of evading immune responses, and cells whose phenotype falls short of satisfying this steady state would expire under hostile immune responses. This view was supported until recently by experimental evidence showing an inferior endurance of severe combined immunodeficiency (SCID)-derived pancreatic islets when engrafted into syngeneic immune-intact wild-type (WT) mice, relative to islets from WT. Here we extend the experimental exploration of ecoimmunity by searching for the presence of the phenotypic changes suggested by the theory. Immune-related phenotypes of islets, spleen, and bone marrow immune cells were determined, as well as SCID and WT nonlymphocytic cells. Islet submass grafting was performed to depict syngeneic graft functionality. Islet cultures were examined under both resting and inflamed conditions for expression of CD40 and major histocompatibility complex (MHC) class I/II and release of interleukin-1α (IL-1α), IL-1β, IL-6, tumor necrosis factor-α (TNF-α), IL-10, and insulin. Results depict multiple pathways that appear to be related to the sculpting of nonimmune cells by immune cells; 59 SCID islet genes displayed relative expression changes compared with WT islets. SCID cells expressed lower tolerability to inflammation and higher levels of immune-related molecules, including MHC class I. Accordingly, islets exhibited a marked increase in insulin release upon immunocyte depletion, in effect resuming endocrine function that was otherwise suppressed by resident immunocytes. This work provides further support of the ecoimmunity theory and encourages subsequent studies to identify its role in the emergence and treatment of autoimmune pathologies, transplant rejection, and cancer.
- Published
- 2016
- Full Text
- View/download PDF
44. Disruption of IKAROS activity in primitive chronic-phase CML cells mimics myeloid disease progression.
- Author
-
Beer PA, Knapp DJ, Miller PH, Kannan N, Sloma I, Heel K, Babovic S, Bulaeva E, Rabu G, Terry J, Druker BJ, Loriaux MM, Loeb KR, Radich JP, Erber WN, and Eaves CJ
- Subjects
- Animals, Antigens, CD34 metabolism, Apoptosis drug effects, Basophils drug effects, Basophils metabolism, Blotting, Western, Cell Proliferation drug effects, Cells, Cultured, Disease Progression, Eosinophils drug effects, Eosinophils metabolism, Flow Cytometry, Humans, Ikaros Transcription Factor genetics, Ikaros Transcription Factor metabolism, Immunoenzyme Techniques, Leukemia, Myeloid, Chronic-Phase genetics, Leukemia, Myeloid, Chronic-Phase metabolism, Mice, Mice, Inbred NOD, Mice, SCID, Protein Kinase Inhibitors pharmacology, Protein-Tyrosine Kinases antagonists & inhibitors, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, STAT5 Transcription Factor genetics, Basophils pathology, Cell Differentiation drug effects, Eosinophils pathology, Ikaros Transcription Factor antagonists & inhibitors, Leukemia, Myeloid, Chronic-Phase pathology, STAT5 Transcription Factor metabolism
- Abstract
Without effective therapy, chronic-phase chronic myeloid leukemia (CP-CML) evolves into an acute leukemia (blast crisis [BC]) that displays either myeloid or B-lymphoid characteristics. This transition is often preceded by a clinically recognized, but biologically poorly characterized, accelerated phase (AP). Here, we report that IKAROS protein is absent or reduced in bone marrow blasts from most CML patients with advanced myeloid disease (AP or BC). This contrasts with primitive CP-CML cells and BCR-ABL1-negative acute myeloid leukemia blasts, which express readily detectable IKAROS. To investigate whether loss of IKAROS contributes to myeloid disease progression in CP-CML, we examined the effects of forced expression of a dominant-negative isoform of IKAROS (IK6) in CP-CML patients' CD34(+) cells. We confirmed that IK6 disrupts IKAROS activity in transduced CP-CML cells and showed that it confers on them features of AP-CML, including a prolonged increased output in vitro and in xenografted mice of primitive cells with an enhanced ability to differentiate into basophils. Expression of IK6 in CD34(+) CP-CML cells also led to activation of signal transducer and activator of transcription 5 and transcriptional repression of its negative regulators. These findings implicate loss of IKAROS as a frequent step and potential diagnostic harbinger of progressive myeloid disease in CML patients., (© 2015 by The American Society of Hematology.)
- Published
- 2015
- Full Text
- View/download PDF
45. Success of interferon α therapy in a patient with hepatitis C virus-negative splenic marginal zone lymphoma associated with polycythemia vera.
- Author
-
Noel N, Sloma I, Aumont C, Hermine O, Turhan A, and Besson C
- Subjects
- Adult, Humans, Immunophenotyping, Janus Kinase 2 genetics, Lymphoma diagnosis, Male, Mutation, Polycythemia Vera diagnosis, Splenic Neoplasms diagnosis, Tomography, X-Ray Computed, Treatment Outcome, Interferon-alpha therapeutic use, Lymphoma complications, Lymphoma drug therapy, Polycythemia Vera complications, Splenic Neoplasms complications, Splenic Neoplasms drug therapy
- Published
- 2015
- Full Text
- View/download PDF
46. Haemophagocytic histiocyte in a peripheral blood film.
- Author
-
Sloma I, Vincent H, Addebbous A, Rivoisy C, Turhan AG, and Michot JM
- Subjects
- Aged, Fatal Outcome, Female, Humans, Lymphohistiocytosis, Hemophagocytic diagnosis, Lymphohistiocytosis, Hemophagocytic drug therapy, Histiocytes pathology, Lymphohistiocytosis, Hemophagocytic blood, Phagocytosis
- Published
- 2014
- Full Text
- View/download PDF
47. Genotypic and functional diversity of phenotypically defined primitive hematopoietic cells in patients with chronic myeloid leukemia.
- Author
-
Sloma I, Beer PA, Saw KM, Chan M, Leung D, Raghuram K, Brimacombe C, Johnston B, Lambie K, Forrest D, Jiang X, and Eaves CJ
- Subjects
- Animals, Bone Marrow Cells cytology, Bone Marrow Transplantation, Cells, Cultured, Female, Fusion Proteins, bcr-abl genetics, Genes, abl genetics, Genotype, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells metabolism, Humans, In Situ Hybridization, Male, Mice, Mice, SCID, Phenotype, Time Factors, Genetic Variation, Hematopoietic Stem Cells pathology, Leukemia, Myeloid, Chronic-Phase genetics, Leukemia, Myeloid, Chronic-Phase pathology
- Abstract
Much progress has been made in the management of chronic-phase chronic myeloid leukemia (CP-CML), but there is a continuing imperative to develop curative treatments, predict patient responses to specific modalities, and anticipate disease relapse or progression. These needs underlie continuing interest in methods to detect and quantify the relevant leukemic cells in clinical samples with improved reliability and specificity. We report the results of comparing three methods to enumerate primitive CP-CML cells in the same samples: genotyping CD34(+)38(-) cells directly by fluorescence in situ hybridization, and measuring BCR-ABL1 transcript-genotyped colony-forming cell outputs in either 5-week long-term cultures (LTCs) containing non-engineered mouse fibroblasts or in 6-week LTCs containing mouse fibroblasts engineered to produce human Steel factor, granulocyte colony-stimulating factor, and IL-3. The results demonstrate that the first two methods significantly overestimate the prevalence of primitive CP-CML cells by comparison to the third. In additional studies, we found that CML-CD34(+) cells can repopulate the marrow and spleen of serially transplanted adult NOD/SCID-IL-2Rγ chain-null mice for more than 1 year with an almost exclusive myeloid differentiation in primary and secondary recipients and without evidence of disease progression. These findings underscore the importance of long-term functional in vitro and in vivo endpoints to identify and characterize CP-CML stem cells., (Crown Copyright © 2013. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
48. IL-27 acts on DCs to suppress the T cell response and autoimmunity by inducing expression of the immunoregulatory molecule CD39.
- Author
-
Mascanfroni ID, Yeste A, Vieira SM, Burns EJ, Patel B, Sloma I, Wu Y, Mayo L, Ben-Hamo R, Efroni S, Kuchroo VK, Robson SC, and Quintana FJ
- Subjects
- Animals, Antibodies immunology, Antigen Presentation drug effects, Antigen Presentation immunology, Antigens, CD metabolism, Apyrase metabolism, Autoantibodies immunology, Carrier Proteins metabolism, Cell Differentiation genetics, Cell Differentiation immunology, Cells, Cultured, Cytokines biosynthesis, Dendritic Cells metabolism, Encephalomyelitis, Autoimmune, Experimental genetics, Encephalomyelitis, Autoimmune, Experimental immunology, Gene Expression, Gene Expression Regulation drug effects, Immune Tolerance immunology, Mice, Mice, Knockout, Myelin Sheath immunology, NLR Family, Pyrin Domain-Containing 3 Protein, Receptors, Cytokine genetics, Receptors, Cytokine metabolism, Receptors, Interleukin, Signal Transduction, T-Lymphocyte Subsets cytology, Transcription, Genetic drug effects, Antigens, CD genetics, Apyrase genetics, Autoimmunity drug effects, Dendritic Cells drug effects, Dendritic Cells immunology, Interleukin-17 pharmacology, T-Lymphocyte Subsets drug effects, T-Lymphocyte Subsets immunology
- Abstract
Dendritic cells (DCs) control the balance between effector T cells and regulatory T cells in vivo. Hence, the study of DCs might identify mechanisms of disease pathogenesis and guide new therapeutic approaches for disorders mediated by the immune system. We found that interleukin 27 (IL-27) signaling in mouse DCs limited the generation of effector cells of the TH1 and TH17 subsets of helper T cells and the development of experimental autoimmune encephalomyelitis (EAE). The effects of IL-27 were mediated at least in part through induction of the immunoregulatory molecule CD39 in DCs. IL-27-induced CD39 decreased the extracellular concentration of ATP and downregulated nucleotide-dependent activation of the NLRP3 inflammasome. Finally, therapeutic vaccination with IL-27-conditioned DCs suppressed established relapsing-remitting EAE. Thus, IL-27 signaling in DCs limited pathogenic T cell responses and the development of autoimmunity.
- Published
- 2013
- Full Text
- View/download PDF
49. Targeting primitive chronic myeloid leukemia cells by effective inhibition of a new AHI-1-BCR-ABL-JAK2 complex.
- Author
-
Chen M, Gallipoli P, DeGeer D, Sloma I, Forrest DL, Chan M, Lai D, Jorgensen H, Ringrose A, Wang HM, Lambie K, Nakamoto H, Saw KM, Turhan A, Arlinghaus R, Paul J, Stobo J, Barnett MJ, Eaves A, Eaves CJ, Holyoake TL, and Jiang X
- Subjects
- Adaptor Proteins, Vesicular Transport, Administration, Oral, Animals, Antigens, CD34 analysis, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Apoptosis drug effects, Benzamides administration & dosage, Biological Availability, Blotting, Western, Cell Proliferation drug effects, DNA Mutational Analysis, Fusion Proteins, bcr-abl genetics, Gene Expression Regulation, Neoplastic, Humans, Imatinib Mesylate, Immunoprecipitation, Mice, Mutation, Neoplastic Stem Cells metabolism, Phosphorylation drug effects, Piperazines administration & dosage, Protein Kinase Inhibitors administration & dosage, Pyrimidines administration & dosage, Remission Induction, Sulfonamides pharmacology, Up-Regulation, Adaptor Proteins, Signal Transducing metabolism, Antineoplastic Combined Chemotherapy Protocols pharmacology, Benzamides pharmacology, Fusion Proteins, bcr-abl antagonists & inhibitors, Fusion Proteins, bcr-abl metabolism, Janus Kinase 2 metabolism, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Leukemia, Myelogenous, Chronic, BCR-ABL Positive metabolism, Microfilament Proteins metabolism, Neoplastic Stem Cells drug effects, Piperazines pharmacology, Protein Kinase Inhibitors pharmacology, Pyrimidines pharmacology
- Abstract
Background: Imatinib mesylate (IM) induces clinical remission of chronic myeloid leukemia (CML). The Abelson helper integration site 1 (AHI-1) oncoprotein interacts with BCR-ABL and Janus kinase 2 (JAK2) to mediate IM response of primitive CML cells, but the effect of the interaction complex on the response to ABL and JAK2 inhibitors is unknown., Methods: The AHI-1-BCR-ABL-JAK2 interaction complex was analyzed by mutational analysis and coimmunoprecipitation. Roles of the complex in regulation of response or resistance to ABL and JAK2 inhibitors were investigated in BCR-ABL (+) cells and primary CML stem/progenitor cells and in immunodeficient NSG mice. All statistical tests were two-sided., Results: The WD40-repeat domain of AHI-1 interacts with BCR-ABL, whereas the N-terminal region interacts with JAK2; loss of these interactions statistically significantly increased the IM sensitivity of CML cells. Disrupting this complex with a combination of IM and an orally bioavailable selective JAK2 inhibitor (TG101209 [TG]) statistically significantly induced death of AHI-1-overexpressing and IM-resistant cells in vitro and enhanced survival of leukemic mice, compared with single agents (combination vs TG alone: 63 vs 53 days, ratio = 0.84, 95% confidence interval [CI] = 0.6 to 1.1, P = .004; vs IM: 57 days, ratio = 0.9, 95% CI = 0.61 to 1.2, P = .003). Combination treatment also statistically significantly enhanced apoptosis of CD34(+) leukemic stem/progenitor cells and eliminated their long-term leukemia-initiating activity in NSG mice. Importantly, this approach was effective against treatment-naive CML stem cells from patients who subsequently proved to be resistant to IM therapy., Conclusions: Simultaneously targeting BCR-ABL and JAK2 activities in CML stem/progenitor cells may improve outcomes in patients destined to develop IM resistance.
- Published
- 2013
- Full Text
- View/download PDF
50. TEL me all.
- Author
-
Sloma I and Eaves CJ
- Subjects
- Animals, Animals, Genetically Modified, Core Binding Factor Alpha 2 Subunit metabolism, Disease Progression, Mice, Models, Animal, Oncogene Fusion, Oncogene Proteins, Fusion metabolism, Oncogenes, Core Binding Factor Alpha 2 Subunit genetics, Leukemia, Myeloid, Acute genetics, Oncogene Proteins, Fusion genetics
- Abstract
Genetically engineered mouse models are powerful systems for dissecting the process of human leukemogenesis. In this issue of Cell Stem Cell, Schindler et al. (2009) describe the elegant use of this approach to dissect, but not completely replicate, the multistep pathogenesis of TEL-AML1(+) leukemia.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.