81 results on '"Quek, Lynn"'
Search Results
2. Molecular Features Associated with Response to Combination Therapy with Enasidenib (ENA) Plus Azacitidine (AZA) in Newly Diagnosed IDH2-Mutated Acute Myeloid Leukemia (AML)
- Author
-
Risueño, Alberto, See, Wendy L., DiNardo, Courtney D., Döhner, Hartmut, Stein, Eytan, Fathi, Amir T., Vyas, Paresh, Quek, Lynn, Prebet, Thomas, Gandhi, Anita K., and Hasan, Maroof
- Published
- 2022
- Full Text
- View/download PDF
3. Therapy-related leukaemias with balanced translocations can arise from pre-existing clonal haematopoiesis
- Author
-
Dillon, Richard, Ahearne, Matthew J., Quek, Lynn, Potter, Nicola, Jovanovic, Jelena, Foot, Nicola, Valganon, Mikel, Jayne, Sandrine, Dennis, Mike, Raj, Kavita, Tauro, Sudhir, Dyer, Martin J. S., Russell, Nigel, Solomon, Ellen, and Grimwade, David
- Published
- 2021
- Full Text
- View/download PDF
4. Integrating clinical features with genetic factors enhances survival prediction for adults with acute myeloid leukemia
- Author
-
Silveira, Douglas R. A., Quek, Lynn, Santos, Itamar S., Corby, Anna, Coelho-Silva, Juan L., Pereira-Martins, Diego A., Vallance, Grant, Brown, Benjamin, Nardinelli, Luciana, Silva, Wellington F., Velloso, Elvira D. R. P., Lucena-Araujo, Antonio R., Traina, Fabiola, Peniket, Andy, Vyas, Paresh, Rego, Eduardo M., Bendit, Israel, and Rocha, Vanderson
- Abstract
The 2017 European LeukemiaNet 2017 acute myeloid leukemia (AML) risk stratification (ELN2017) is widely used for risk-stratifying patients with AML. However, its applicability in low- and middle-income countries is limited because of a lack of full cytogenetic and molecular information at diagnosis. Here, we propose an alternative for risk stratification (the Adapted Genetic Risk [AGR]), which permits cytogenetic or molecular missing data while retaining prognostic power. We first analyzed 167 intensively treated patients with nonacute promyelocytic leukemia AML enrolled in São Paulo, Brazil (Faculdade de Medicina da Universidade de São Paulo), as our training data set, using ELN2017 as the standard for comparison with our AGR. Next, we combined our AGR with clinical prognostic parameters found in a Cox proportional hazards model to create a novel scoring system (survival AML score, SAMLS) that stratifies patients with newly diagnosed AML. Finally, we have used 2 independent test cohorts, Faculdade de Medicina de Ribeirão Preto (FMRP; Brazil, n = 145) and Oxford University Hospitals (OUH; United Kingdom, n = 157) for validating our findings. AGR was statistically significant for overall survival (OS) in both test cohorts (FMRP, P = .037; OUH, P = .012) and disease-free survival in FMRP (P = .04). The clinical prognostic features in SAMLS were age (>45 years), white blood cell count (<1.5 or >30.0 × 103/μL), and low albumin levels (<3.8 g/dL), which were associated with worse OS in all 3 cohorts. SAMLS showed a significant difference in OS in the training cohort (P < .001) and test cohorts (FMRP, P = .0018; OUH, P < .001). Therefore, SAMLS, which incorporates the novel AGR evaluation with clinical parameters, is an accurate tool for AML risk assessment.
- Published
- 2020
- Full Text
- View/download PDF
5. Integrating clinical features with genetic factors enhances survival prediction for adults with acute myeloid leukemia
- Author
-
Silveira, Douglas R.A., Quek, Lynn, Santos, Itamar S., Corby, Anna, Coelho-Silva, Juan L., Pereira-Martins, Diego A., Vallance, Grant, Brown, Benjamin, Nardinelli, Luciana, Silva, Wellington F., Velloso, Elvira D.R.P., Lucena-Araujo, Antonio R., Traina, Fabiola, Peniket, Andy, Vyas, Paresh, Rego, Eduardo M., Bendit, Israel, and Rocha, Vanderson
- Abstract
The 2017 European LeukemiaNet 2017 acute myeloid leukemia (AML) risk stratification (ELN2017) is widely used for risk-stratifying patients with AML. However, its applicability in low- and middle-income countries is limited because of a lack of full cytogenetic and molecular information at diagnosis. Here, we propose an alternative for risk stratification (the Adapted Genetic Risk [AGR]), which permits cytogenetic or molecular missing data while retaining prognostic power. We first analyzed 167 intensively treated patients with nonacute promyelocytic leukemia AML enrolled in São Paulo, Brazil (Faculdade de Medicina da Universidade de São Paulo), as our training data set, using ELN2017 as the standard for comparison with our AGR. Next, we combined our AGR with clinical prognostic parameters found in a Cox proportional hazards model to create a novel scoring system (survival AML score, SAMLS) that stratifies patients with newly diagnosed AML. Finally, we have used 2 independent test cohorts, Faculdade de Medicina de Ribeirão Preto (FMRP; Brazil, n = 145) and Oxford University Hospitals (OUH; United Kingdom, n = 157) for validating our findings. AGR was statistically significant for overall survival (OS) in both test cohorts (FMRP, P= .037; OUH, P= .012) and disease-free survival in FMRP (P= .04). The clinical prognostic features in SAMLS were age (>45 years), white blood cell count (<1.5 or >30.0 × 103/μL), and low albumin levels (<3.8 g/dL), which were associated with worse OS in all 3 cohorts. SAMLS showed a significant difference in OS in the training cohort (P< .001) and test cohorts (FMRP, P= .0018; OUH, P< .001). Therefore, SAMLS, which incorporates the novel AGR evaluation with clinical parameters, is an accurate tool for AML risk assessment.
- Published
- 2020
- Full Text
- View/download PDF
6. Molecular mechanisms mediating relapse following ivosidenib monotherapy in IDH1-mutant relapsed or refractory AML
- Author
-
Choe, Sung, Wang, Hongfang, DiNardo, Courtney D., Stein, Eytan M., de Botton, Stéphane, Roboz, Gail J., Altman, Jessica K., Mims, Alice S., Watts, Justin M., Pollyea, Daniel A., Fathi, Amir T., Tallman, Martin S., Kantarjian, Hagop M., Stone, Richard M., Quek, Lynn, Konteatis, Zenon, Dang, Lenny, Nicolay, Brandon, Nejad, Parham, Liu, Guowen, Zhang, Vickie, Liu, Hua, Goldwasser, Meredith, Liu, Wei, Marks, Kevin, Bowden, Chris, Biller, Scott A., Attar, Eyal C., and Wu, Bin
- Abstract
Isocitrate dehydrogenase (IDH) 1 and 2 mutations result in overproduction of D-2-hydroxyglutarate (2-HG) and impaired cellular differentiation. Ivosidenib, a targeted mutant IDH1 (mIDH1) enzyme inhibitor, can restore normal differentiation and results in clinical responses in a subset of patients with mIDH1 relapsed/refractory (R/R) acute myeloid leukemia (AML). We explored mechanisms of ivosidenib resistance in 174 patients with confirmed mIDH1 R/R AML from a phase 1 trial. Receptor tyrosine kinase (RTK) pathway mutations were associated with primary resistance to ivosidenib. Multiple mechanisms contributed to acquired resistance, particularly outgrowth of RTK pathway mutations and 2-HG–restoring mutations (second-site IDH1 mutations, IDH2 mutations). Observation of multiple concurrent mechanisms in individual patients underscores the complex biology of resistance and has important implications for rational combination therapy design. This trial was registered at www.clinicaltrials.gov as #NCT02074839
- Published
- 2020
- Full Text
- View/download PDF
7. Molecular mechanisms mediating relapse following ivosidenib monotherapy in IDH1-mutant relapsed or refractory AML
- Author
-
Choe, Sung, Wang, Hongfang, DiNardo, Courtney D., Stein, Eytan M., de Botton, Stéphane, Roboz, Gail J., Altman, Jessica K., Mims, Alice S., Watts, Justin M., Pollyea, Daniel A., Fathi, Amir T., Tallman, Martin S., Kantarjian, Hagop M., Stone, Richard M., Quek, Lynn, Konteatis, Zenon, Dang, Lenny, Nicolay, Brandon, Nejad, Parham, Liu, Guowen, Zhang, Vickie, Liu, Hua, Goldwasser, Meredith, Liu, Wei, Marks, Kevin, Bowden, Chris, Biller, Scott A., Attar, Eyal C., and Wu, Bin
- Abstract
Isocitrate dehydrogenase (IDH) 1 and 2 mutations result in overproduction of D-2-hydroxyglutarate (2-HG) and impaired cellular differentiation. Ivosidenib, a targeted mutant IDH1 (mIDH1) enzyme inhibitor, can restore normal differentiation and results in clinical responses in a subset of patients with mIDH1relapsed/refractory (R/R) acute myeloid leukemia (AML). We explored mechanisms of ivosidenib resistance in 174 patients with confirmed mIDH1R/R AML from a phase 1 trial. Receptor tyrosine kinase (RTK) pathway mutations were associated with primary resistance to ivosidenib. Multiple mechanisms contributed to acquired resistance, particularly outgrowth of RTK pathway mutations and 2-HG–restoring mutations (second-site IDH1mutations, IDH2mutations). Observation of multiple concurrent mechanisms in individual patients underscores the complex biology of resistance and has important implications for rational combination therapy design. This trial was registered at www.clinicaltrials.govas #NCT02074839
- Published
- 2020
- Full Text
- View/download PDF
8. Co-occurrence of DNMT3A, NPM1, FLT3 mutations identifies a subset of acute myeloid leukemia with adverse prognosis
- Author
-
Bezerra, Matheus F., Lima, Aleide S., Piqué-Borràs, Maria-Riera, Silveira, Douglas R., Coelho-Silva, Juan L., Pereira-Martins, Diego A., Weinhäuser, Isabel, Franca-Neto, Pedro L., Quek, Lynn, Corby, Anna, Oliveira, Mayara M., Lima, Marinus M., de Assis, Reijane A., de Melo Campos, Paula, Duarte, Bruno K., Bendit, Israel, Rocha, Vanderson, Rego, Eduardo M., Traina, Fabiola, Saad, Sara T., Beltrão, Eduardo I., Bezerra, Marcos A., and Lucena-Araujo, Antonio R.
- Published
- 2020
- Full Text
- View/download PDF
9. Co-occurrence of DNMT3A, NPM1, FLT3mutations identifies a subset of acute myeloid leukemia with adverse prognosis
- Author
-
Bezerra, Matheus F., Lima, Aleide S., Piqué-Borràs, Maria-Riera, Silveira, Douglas R., Coelho-Silva, Juan L., Pereira-Martins, Diego A., Weinhäuser, Isabel, Franca-Neto, Pedro L., Quek, Lynn, Corby, Anna, Oliveira, Mayara M., Lima, Marinus M., de Assis, Reijane A., de Melo Campos, Paula, Duarte, Bruno K., Bendit, Israel, Rocha, Vanderson, Rego, Eduardo M., Traina, Fabiola, Saad, Sara T., Beltrão, Eduardo I., Bezerra, Marcos A., and Lucena-Araujo, Antonio R.
- Published
- 2020
- Full Text
- View/download PDF
10. TP53mutation status divides myelodysplastic syndromes with complex karyotypes into distinct prognostic subgroups
- Author
-
Haase, Detlef, Stevenson, Kristen E., Neuberg, Donna, Maciejewski, Jaroslaw P., Nazha, Aziz, Sekeres, Mikkael A., Ebert, Benjamin L., Garcia-Manero, Guillermo, Haferlach, Claudia, Haferlach, Torsten, Kern, Wolfgang, Ogawa, Seishi, Nagata, Yasunobu, Yoshida, Kenichi, Graubert, Timothy A., Walter, Matthew J., List, Alan F., Komrokji, Rami S., Padron, Eric, Sallman, David, Papaemmanuil, Elli, Campbell, Peter J., Savona, Michael R., Seegmiller, Adam, Adès, Lionel, Fenaux, Pierre, Shih, Lee-Yung, Bowen, David, Groves, Michael J., Tauro, Sudhir, Fontenay, Michaela, Kosmider, Olivier, Bar-Natan, Michal, Steensma, David, Stone, Richard, Heuser, Michael, Thol, Felicitas, Cazzola, Mario, Malcovati, Luca, Karsan, Aly, Ganster, Christina, Hellström-Lindberg, Eva, Boultwood, Jacqueline, Pellagatti, Andrea, Santini, Valeria, Quek, Lynn, Vyas, Paresh, Tüchler, Heinz, Greenberg, Peter L., and Bejar, Rafael
- Abstract
Risk stratification is critical in the care of patients with myelodysplastic syndromes (MDS). Approximately 10% have a complex karyotype (CK), defined as more than two cytogenetic abnormalities, which is a highly adverse prognostic marker. However, CK-MDS can carry a wide range of chromosomal abnormalities and somatic mutations. To refine risk stratification of CK-MDS patients, we examined data from 359 CK-MDS patients shared by the International Working Group for MDS. Mutations were underrepresented with the exception of TP53mutations, identified in 55% of patients. TP53mutated patients had even fewer co-mutated genes but were enriched for the del(5q) chromosomal abnormality (p< 0.005), monosomal karyotype (p< 0.001), and high complexity, defined as more than 4 cytogenetic abnormalities (p< 0.001). Monosomal karyotype, high complexity, and TP53mutation were individually associated with shorter overall survival, but monosomal status was not significant in a multivariable model. Multivariable survival modeling identified severe anemia (hemoglobin < 8.0 g/dL), NRASmutation, SF3B1mutation, TP53mutation, elevated blast percentage (>10%), abnormal 3q, abnormal 9, and monosomy 7 as having the greatest survival risk. The poor risk associated with CK-MDS is driven by its association with prognostically adverse TP53mutations and can be refined by considering clinical and karyotype features.
- Published
- 2019
- Full Text
- View/download PDF
11. Clonal heterogeneity of acute myeloid leukemia treated with the IDH2 inhibitor enasidenib
- Author
-
Quek, Lynn, David, Muriel D., Kennedy, Alison, Metzner, Marlen, Amatangelo, Michael, Shih, Alan, Stoilova, Bilyana, Quivoron, Cyril, Heiblig, Maël, Willekens, Christophe, Saada, Véronique, Alsafadi, Samar, Vijayabaskar, M. S., Peniket, Andy, Bernard, Oliver A., Agresta, Sam, Yen, Katharine, MacBeth, Kyle, Stein, Eytan, Vassiliou, George S., Levine, Ross, De Botton, Stephane, Thakurta, Anjan, Penard-Lacronique, Virginie, and Vyas, Paresh
- Abstract
Mutations in the gene encoding isocitrate dehydrogenase 2 (IDH2) occur in several types of cancer, including acute myeloid leukemia (AML). In model systems, mutant IDH2 causes hematopoietic differentiation arrest. Enasidenib, a selective small-molecule inhibitor of mutant IDH2, produces a clinical response in 40% of treated patients with relapsed/refractory AML by promoting leukemic cell differentiation. Here, we studied the clonal basis of response and acquired resistance to enasidenib treatment. Using sequential patient samples, we determined the clonal structure of hematopoietic cell populations at different stages of differentiation. Before therapy, IDH2-mutant clones showed variable differentiation arrest. Enasidenib treatment promoted hematopoietic differentiation from either terminal or ancestral mutant clones; less frequently, treatment promoted differentiation of nonmutant cells. Analysis of paired diagnosis/relapse samples did not identify second-site mutations in IDH2at relapse. Instead, relapse arose by clonal evolution or selection of terminal or ancestral clones, thus highlighting multiple bypass pathways that could potentially be targeted to restore differentiation arrest. These results show how mapping of clonal structure in cell populations at different stages of differentiation can reveal the response and evolution of clones during treatment response and relapse.
- Published
- 2018
- Full Text
- View/download PDF
12. An Integrated Single Cell Immunogenomic Atlas Reveals Pathways of Clonal Expansion, Inflammation and Dyserythropoiesis in Low and High-Risk Clonal Haematopoiesis
- Author
-
Zheng, Xuesen, Tan, Amandine, Casares Alaez, Maria del Pilar, Mallett, Garry, Kaya, Deniz Ece, Seymen, Nogayhan, Rzepkowska, Marta, Mackie, Sarah, Chatzikyriakou, Prodromos, Bogaanay, Laarni, Lewis, Jen, Hasan, Maroof, Suragani, Rajasekhar N.V.S., Carmichael, James, Mufti, Ghulam, Gandhi, Anita K., Napolitani, Giorgio, and Quek, Lynn
- Abstract
Clonal haematopoiesis (CH) describes age-related clonal expansion of mutant haematopoietic cells in the absence of overt haematological disease. Growing evidence suggests that individuals with ‘high risk’ (HR-CH) have increased risk of blood cancer, auto-inflammatory and cerebro-cardiovascular disease. We aimed to better understand genomic and immunological events associated with progression of CH. By setting up a cohort of individuals undergoing hip arthroplasty ( n=432, 58% female, age range 50-90, mean 69 years), we detected CH mutations (sensitivity to 0.2% variant allele frequency, VAF) in 38 % of individuals. We studied their haematopoietic and immune landscape to gain deeper insights into the impact of CH.
- Published
- 2023
- Full Text
- View/download PDF
13. CEBPA-Driven Expression of the Transcriptionally Inactive deltaTP73 Isoform Phenocopies TP53mutated Poor Risk and Drug-Resistant Acute Myeloid Leukemia
- Author
-
Pereira-Martins, Diego, Ortiz Rojas, César Alexander, Weinhaeuser, Isabel, Wierenga, Bart-Jan, Van Den Boom, Vincent, Mojallali, Fatemeh, Sternadt, Dominique, Bianco, Thiago Mantello, van der Meer, Nisha, Silveira, Douglas RA, Quek, Lynn, Ammatuna, Emanuele, Lucena-Araujo, Antonio R, Huls, Gerwin A., Rego, Eduardo Magalhães, and Schuringa, Jan Jacob
- Abstract
Mutations in TP53( TP53mut) are present in roughly 10% of acute myeloid leukemia (AML) patients and represent a unique subgroup of patients with very poor prognosis. While the exact mechanisms of TP53mut-driven leukemogenesis remain elusive, transcriptional changes associated with suppression of the TP53 signaling pathway are considered to confer a dismal prognosis on TP53mut patients. Using molecular profiling of a large cohort of AML patients (n=823) we identify a new poor prognostic subgroup in AML that is TP53 WTbut shares strong similarities with TP53mut patients. Patients included in this new group present increased expression of the oncogenic TP73gene isoform ΔNp73,which lacks the transactivation domain but still binds to chromatin. We find that the transcriptional and metabolic program of ΔNp73highAMLs strongly resembles that of TP53mut AML, being enriched for stemness signatures like “17LSC” and “LSC UP”. In line with TP53mut AML, ΔNp73high/ TP53WTfrequently co-occurs with U2AF1, SRSF2, TET2, and RUNX1mutations .Lentiviral overexpression (OE) of ΔNp73 in healthy CD34 +cells increased cell proliferation and stemness. ΔNp73-OE in TP53WTAML models, imposed drug resistance against several standard-of-care cytotoxic therapies in AML (e.g., AraC, venetoclax (VEN) ± azacytidine (AZA), and FLT3 inhibitors), while drug resistance was not further enhanced upon ΔNp73-OE in TP53mut AML models. These results were validated using ex vivotreated primary AML samples (n=80), whereby ΔNp73 highsamples were more resistant to several cytotoxic therapies, including VEN+AZA .Lentiviral ΔNp73-OE in primary APL samples (n=10) allowed efficient engraftment in vivousing xenograft mouse models with colonization of secondary organs and splenomegaly, which was not observed in the empty vector controls.
- Published
- 2023
- Full Text
- View/download PDF
14. Metformin-Induced Ferroptosis Is a Therapeutic Vulnerability in IDH2-Mutant AML Linked to Metabolic Rewiring Towards Fatty Acid Oxidation
- Author
-
Sternadt, Dominique, Pereira-Martins, Diego, Silveira, Douglas RA, Weinhaeuser, Isabel, Yang, Ming, Chatzikyriakou, Prodromos, Casares Alaez, Pilar, Ammatuna, Emanuele, Oudejans, Lieve, Huls, Gerwin A., Frezza, Christian, Quek, Lynn, and Schuringa, Jan Jacob
- Abstract
Metabolic rewiring is an essential feature of leukemic cells to sustain tumorigenesis and is largely influenced by mutational status. We previously demonstrated this phenomenon in FLT3-ITD acute myeloid leukemia (AML) upon combined inhibition of complex II activity and lactate import (Erdem et al., 2022). Mutations in isocitrate dehydrogenase ( IDH1/2mut) occur in about 20% of AML cases, and despite the emergence of targeted therapies, patients often relapse. The molecular and epigenetic outcomes of 2-R-hydroxyglutarate accumulation and competitive inhibition of histone demethylases have been extensively studied in IDH1/2mutAMLs, but the metabolic consequences are still largely unexplored. Here, we applied multi-omics studies on cell line models and primary AML samples combined with functional studies to unravel the metabolic rewiring of IDH1/2mutAMLs.
- Published
- 2023
- Full Text
- View/download PDF
15. Metabolism-Related Features Identify the Combination Metformin Plus NAMPT Inhibitors As a Selective Treatment Strategy in Acute Myeloid Leukemia
- Author
-
Weinhaeuser, Isabel, Pereira-Martins, Diego, Sternadt, Dominique, Griessinger, Emmanuel F, Silveira, Douglas RA, Coelho-Silva, Juan L, Alves-Silva, Antonio Bruno, Erdem, Ayşegül, Duarte, Bruno Koza Lino, Quek, Lynn, Traina, Fabiola, Saad, Sara Teresinha Olalla, Hilberink, Jacobien R, Ammatuna, Emanuele, Rego, Eduardo Magalhães, Huls, Geert A., Lucena-Araujo, Antonio R, and Schuringa, Jan Jacob
- Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease, which remains difficult to treat due to its heterogeneous nature. Recently, metabolic adaptation has been recognized as a trademark of leukemogenesis. Notably, altered function of mitochondria (mt) in leukemic stem cells (LSCs) has been linked to chemoresistance in some patients, causing relapse of disease. Thus, targeting the rewired energy metabolism of leukemic cells has gained a lot of attention in recent years.
- Published
- 2023
- Full Text
- View/download PDF
16. Impaired Metabolic Plasticity Under Stress Constitutes a Therapeutic Vulnerability in IDH1/2-Mutant Acute Myeloid Leukemia
- Author
-
Sternadt, Dominique, Pereira-Martins, Diego, Silveira, Douglas RA, Chatzikyriakou, Prodromos, Casares Alaez, Maria del Pilar, Weinhaeuser, Isabel, Hogeling, Shanna M, Ammatuna, Emanuele, Huls, Gerwin A., Quek, Lynn, and Schuringa, Jan Jacob
- Published
- 2022
- Full Text
- View/download PDF
17. High MN1 Expression Is Associated with an Lspc-Enriched Phenotype and Glycolysis Representing a New Vulnerability in Acute Myeloid Leukemia
- Author
-
Pereira-Martins, Diego, van Dijk, Noortje, Weinhaeuser, Isabel, Silveira, Douglas RA, Coelho Da Silva, Juan, Bianco, Thiago M, Ortiz, César Alexander, Traina, Fabiola, Figueiredo-Pontes, Lorena L, Ammatuna, Emanuele, Quek, Lynn, Huls, Gerwin A., Lucena-Araujo, Antonio R., Rego, Eduardo M, and Schuringa, Jan Jacob
- Published
- 2022
- Full Text
- View/download PDF
18. Single-Cell Transcriptome Analysis Reveals Pathways That Drive Dyserythropoiesis in Myelodysplastic Syndromes, Which Are Targeted By Luspatercept
- Author
-
Chowdhury, Onima, Silveira, Douglas RA, Reed, Sophie, O'Sullivan, Jennifer, Clark, Sally-Ann, Ashley, Neil, Psaila, Bethan, Thongjuea, Supat, Mead, Adam J, and Quek, Lynn
- Published
- 2022
- Full Text
- View/download PDF
19. Dysregulation of Stem-Progenitor and Differentiation Programmes through Modulation of Bivalent Chromatin Drives Leukemogenesis in Mutant IDH2 Acute Myeloid Leukaemia
- Author
-
Silveira, Douglas RA RA, Chatzikyriakou, Prodromos, Casares Alaez, Maria del Pilar, Mackie, Sarah, Hulks, Roan, Yavorska, Olena, Siejka-Zielinska, Paulina, Metzner, Marlen, Smith, Alastair, Usukhbayar, Batchimeg, Penard-Lacronique, Virginie, Song, Chunxiao, Gandhi, Anita K., Hasan, Maroof, Vyas, Paresh, Milne, Thomas A., Kriaucionis, Skirmantas, and Quek, Lynn
- Published
- 2022
- Full Text
- View/download PDF
20. Single-Cell Transcriptome Analysis Reveals Pathways That Drive Dyserythropoiesis in Myelodysplastic Syndromes, Which Are Targeted By Luspatercept
- Author
-
Chowdhury, Onima, Silveira, Douglas RA, Reed, Sophie, O'Sullivan, Jennifer, Clark, Sally-Ann, Ashley, Neil, Psaila, Bethan, Thongjuea, Supat, Mead, Adam J, and Quek, Lynn
- Published
- 2022
- Full Text
- View/download PDF
21. Dysregulation of Stem-Progenitor and Differentiation Programmes through Modulation of Bivalent Chromatin Drives Leukemogenesis in Mutant IDH2 Acute Myeloid Leukaemia
- Author
-
Silveira, Douglas RA RA, Chatzikyriakou, Prodromos, Casares Alaez, Maria del Pilar, Mackie, Sarah, Hulks, Roan, Yavorska, Olena, Siejka-Zielinska, Paulina, Metzner, Marlen, Smith, Alastair, Usukhbayar, Batchimeg, Penard-Lacronique, Virginie, Song, Chunxiao, Gandhi, Anita K., Hasan, Maroof, Vyas, Paresh, Milne, Thomas A., Kriaucionis, Skirmantas, and Quek, Lynn
- Published
- 2022
- Full Text
- View/download PDF
22. Single Cell Characterisation of Transcriptional Programmes of Responsive and Resistant Leukaemic Progenitors in IDH2-Mutant Acute Myeloid Leukaemia in the AG221-AML-005 Study
- Author
-
Chatzikyriakou, Prodromos, Silveira, Douglas RA, Casares Aláez, Maria del Pilar, Usukhbayar, Batchimeg, Metzner, Marlen, Vyas, Paresh, Gandhi, Anita K., Risueño, Alberto, Hasan, Maroof, and Quek, Lynn
- Published
- 2022
- Full Text
- View/download PDF
23. M2 Macrophages Drive Resistance to Phagocytosis and Improve Mitochondrial Metabolism in Acute Myeloid Leukemia Facilitating Leukemic Transformation and In Vivo Engraftment
- Author
-
Weinhaeuser, Isabel, Pereira-Martins, Diego, Yamamoto de Almeida, Luciana, Hilberink, Jacobien R, Silveira, Douglas RA, Quek, Lynn, Ortiz, César Alexander, Lucena-Araujo, Antonio R., Visser, Nienke, Ammatuna, Emanuele, Huls, Gerwin A., Rego, Eduardo M, and Schuringa, Jan Jacob
- Published
- 2022
- Full Text
- View/download PDF
24. Single Cell Characterisation of Transcriptional Programmes of Responsive and Resistant Leukaemic Progenitors in IDH2-Mutant Acute Myeloid Leukaemia in the AG221-AML-005 Study
- Author
-
Chatzikyriakou, Prodromos, Silveira, Douglas RA, Casares Aláez, Maria del Pilar, Usukhbayar, Batchimeg, Metzner, Marlen, Vyas, Paresh, Gandhi, Anita K., Risueño, Alberto, Hasan, Maroof, and Quek, Lynn
- Published
- 2022
- Full Text
- View/download PDF
25. M2 Macrophages Drive Resistance to Phagocytosis and Improve Mitochondrial Metabolism in Acute Myeloid Leukemia Facilitating Leukemic Transformation and In VivoEngraftment
- Author
-
Weinhaeuser, Isabel, Pereira-Martins, Diego, Yamamoto de Almeida, Luciana, Hilberink, Jacobien R, Silveira, Douglas RA, Quek, Lynn, Ortiz, César Alexander, Lucena-Araujo, Antonio R., Visser, Nienke, Ammatuna, Emanuele, Huls, Gerwin A., Rego, Eduardo M, and Schuringa, Jan Jacob
- Published
- 2022
- Full Text
- View/download PDF
26. High MN1 Expression Is Associated with an Lspc-Enriched Phenotype and Glycolysis Representing a New Vulnerability in Acute Myeloid Leukemia
- Author
-
Pereira-Martins, Diego, van Dijk, Noortje, Weinhaeuser, Isabel, Silveira, Douglas RA, Coelho Da Silva, Juan, Bianco, Thiago M, Ortiz, César Alexander, Traina, Fabiola, Figueiredo-Pontes, Lorena L, Ammatuna, Emanuele, Quek, Lynn, Huls, Gerwin A., Lucena-Araujo, Antonio R., Rego, Eduardo M, and Schuringa, Jan Jacob
- Published
- 2022
- Full Text
- View/download PDF
27. Impaired Metabolic Plasticity Under Stress Constitutes a Therapeutic Vulnerability in IDH1/2-Mutant Acute Myeloid Leukemia
- Author
-
Sternadt, Dominique, Pereira-Martins, Diego, Silveira, Douglas RA, Chatzikyriakou, Prodromos, Casares Alaez, Maria del Pilar, Weinhaeuser, Isabel, Hogeling, Shanna M, Ammatuna, Emanuele, Huls, Gerwin A., Quek, Lynn, and Schuringa, Jan Jacob
- Published
- 2022
- Full Text
- View/download PDF
28. Single-cell analysis reveals the continuum of human lympho-myeloid progenitor cells
- Author
-
Karamitros, Dimitris, Stoilova, Bilyana, Aboukhalil, Zahra, Hamey, Fiona, Reinisch, Andreas, Samitsch, Marina, Quek, Lynn, Otto, Georg, Repapi, Emmanouela, Doondeea, Jessica, Usukhbayar, Batchimeg, Calvo, Julien, Taylor, Stephen, Goardon, Nicolas, Six, Emmanuelle, Pflumio, Francoise, Porcher, Catherine, Majeti, Ravindra, Göttgens, Berthold, and Vyas, Paresh
- Abstract
The hierarchy of human hemopoietic progenitor cells that produce lymphoid and granulocytic–monocytic (myeloid) lineages is unclear. Multiple progenitor populations produce lymphoid and myeloid cells, but they remain incompletely characterized. Here we demonstrated that lympho-myeloid progenitor populations in cord blood — lymphoid-primed multi-potential progenitors (LMPPs), granulocyte-macrophage progenitors (GMPs) and multi-lymphoid progenitors (MLPs) — were functionally and transcriptionally distinct and heterogeneous at the clonal level, with progenitors of many different functional potentials present. Although most progenitors had the potential to develop into only one mature cell type (‘uni-lineage potential’), bi- and rarer multi-lineage progenitors were present among LMPPs, GMPs and MLPs. Those findings, coupled with single-cell expression analyses, suggest that a continuum of progenitors execute lymphoid and myeloid differentiation, rather than only uni-lineage progenitors’ being present downstream of stem cells. Vyas and colleagues show that a continuum of hemopoietic progenitor cells from human cord blood execute lymphoid and myeloid differentiation.
- Published
- 2018
- Full Text
- View/download PDF
29. Enasidenib induces acute myeloid leukemia cell differentiation to promote clinical response
- Author
-
Amatangelo, Michael D., Quek, Lynn, Shih, Alan, Stein, Eytan M., Roshal, Mikhail, David, Muriel D., Marteyn, Benoit, Farnoud, Noushin Rahnamay, de Botton, Stephane, Bernard, Olivier A., Wu, Bin, Yen, Katharine E., Tallman, Martin S., Papaemmanuil, Elli, Penard-Lacronique, Virginie, Thakurta, Anjan, Vyas, Paresh, and Levine, Ross L.
- Abstract
Recurrent mutations at R140 and R172 in isocitrate dehydrogenase 2 (IDH2) occur in many cancers, including ∼12% of acute myeloid leukemia (AML). In preclinical models these mutations cause accumulation of the oncogenic metabolite R-2-hydroxyglutarate (2-HG) and induce hematopoietic differentiation block. Single-agent enasidenib (AG-221/CC-90007), a selective mutant IDH2 (mIDH2) inhibitor, produced an overall response rate of 40.3% in relapsed/refractory AML (rrAML) patients with mIDH2 in a phase 1 trial. However, its mechanism of action and biomarkers associated with response remain unclear. Here, we measured 2-HG, mIDH2 allele burden, and co-occurring somatic mutations in sequential patient samples from the clinical trial and correlated these with clinical response. Furthermore, we used flow cytometry to assess inhibition of mIDH2 on hematopoietic differentiation. We observed potent 2-HG suppression in both R140 and R172 mIDH2 AML subtypes, with different kinetics, which preceded clinical response. Suppression of 2-HG alone did not predict response, because most nonresponding patients also exhibited 2-HG suppression. Complete remission (CR) with persistence of mIDH2 and normalization of hematopoietic stem and progenitor compartments with emergence of functional mIDH2 neutrophils were observed. In a subset of CR patients, mIDH2 allele burden was reduced and remained undetectable with response. Co-occurring mutations in NRAS and other MAPK pathway effectors were enriched in nonresponding patients, consistent with RAS signaling contributing to primary therapeutic resistance. Together, these data support differentiation as the main mechanism of enasidenib efficacy in relapsed/refractory AML patients and provide insight into resistance mechanisms to inform future mechanism-based combination treatment studies.
- Published
- 2017
- Full Text
- View/download PDF
30. Enasidenib induces acute myeloid leukemia cell differentiation to promote clinical response
- Author
-
Amatangelo, Michael D., Quek, Lynn, Shih, Alan, Stein, Eytan M., Roshal, Mikhail, David, Muriel D., Marteyn, Benoit, Farnoud, Noushin Rahnamay, de Botton, Stephane, Bernard, Olivier A., Wu, Bin, Yen, Katharine E., Tallman, Martin S., Papaemmanuil, Elli, Penard-Lacronique, Virginie, Thakurta, Anjan, Vyas, Paresh, and Levine, Ross L.
- Abstract
Recurrent mutations at R140 and R172 in isocitrate dehydrogenase 2 (IDH2) occur in many cancers, including ∼12% of acute myeloid leukemia (AML). In preclinical models these mutations cause accumulation of the oncogenic metabolite R-2-hydroxyglutarate (2-HG) and induce hematopoietic differentiation block. Single-agent enasidenib (AG-221/CC-90007), a selective mutant IDH2 (mIDH2) inhibitor, produced an overall response rate of 40.3% in relapsed/refractory AML (rrAML) patients with mIDH2in a phase 1 trial. However, its mechanism of action and biomarkers associated with response remain unclear. Here, we measured 2-HG, mIDH2allele burden, and co-occurring somatic mutations in sequential patient samples from the clinical trial and correlated these with clinical response. Furthermore, we used flow cytometry to assess inhibition of mIDH2 on hematopoietic differentiation. We observed potent 2-HG suppression in both R140 and R172 mIDH2AML subtypes, with different kinetics, which preceded clinical response. Suppression of 2-HG alone did not predict response, because most nonresponding patients also exhibited 2-HG suppression. Complete remission (CR) with persistence of mIDH2and normalization of hematopoietic stem and progenitor compartments with emergence of functional mIDH2neutrophils were observed. In a subset of CR patients, mIDH2allele burden was reduced and remained undetectable with response. Co-occurring mutations in NRAS and other MAPK pathway effectors were enriched in nonresponding patients, consistent with RAS signaling contributing to primary therapeutic resistance. Together, these data support differentiation as the main mechanism of enasidenib efficacy in relapsed/refractory AML patients and provide insight into resistance mechanisms to inform future mechanism-based combination treatment studies.
- Published
- 2017
- Full Text
- View/download PDF
31. Mutational analysis of disease relapse in patients allografted for acute myeloid leukemia
- Author
-
Quek, Lynn, Ferguson, Paul, Metzner, Marlen, Ahmed, Ikhlaaq, Kennedy, Alison, Garnett, Catherine, Jeffries, Sally, Walter, Claudia, Piechocki, Kim, Timbs, Adele, Danby, Robert, Raghavan, Manoj, Peniket, Andrew, Griffiths, Mike, Bacon, Andrew, Ward, Janice, Wheatley, Keith, Vyas, Paresh, and Craddock, Charles
- Abstract
Disease relapse is the major cause of treatment failure after allogeneic stem cell transplantation (allo-SCT) in acute myeloid leukemia (AML). To identify AML-associated genes prognostic of AML relapse post–allo-SCT, we resequenced 35 genes in 113 adults at diagnosis, 49 of whom relapsed. Two hundred sixty-two mutations were detected in 102/113 (90%) patients. An increased risk of relapse was observed in patients with mutations in WT1(P= .018), DNMT3A(P= .045), FLT3 ITD(P= .071), and TP53(P= .06), whereas mutations in IDH1were associated with a reduced risk of disease relapse (P= .018). In 29 patients, we additionally compared mutational profiles in bone marrow at diagnosis and relapse to study changes in clonal structure at relapse. In 13/29 patients, mutational profiles altered at relapse. In 9 patients, mutations present at relapse were not detected at diagnosis. In 15 patients, additional available pre–allo-SCT samples demonstrated that mutations identified posttransplant but not at diagnosis were detectable immediately prior to transplant in 2 of 15 patients. Taken together, these observations, if confirmed in larger studies, have the potential to inform the design of novel strategies to reduce posttransplant relapse highlighting the potential importance of post–allo-SCT interventions with a broad antitumor specificity in contrast to targeted therapies based on mutational profile at diagnosis.
- Published
- 2016
- Full Text
- View/download PDF
32. Mutational analysis of disease relapse in patients allografted for acute myeloid leukemia
- Author
-
Quek, Lynn, Ferguson, Paul, Metzner, Marlen, Ahmed, Ikhlaaq, Kennedy, Alison, Garnett, Catherine, Jeffries, Sally, Walter, Claudia, Piechocki, Kim, Timbs, Adele, Danby, Robert, Raghavan, Manoj, Peniket, Andrew, Griffiths, Mike, Bacon, Andrew, Ward, Janice, Wheatley, Keith, Vyas, Paresh, and Craddock, Charles
- Abstract
Disease relapse is the major cause of treatment failure after allogeneic stem cell transplantation (allo-SCT) in acute myeloid leukemia (AML). To identify AML-associated genes prognostic of AML relapse post–allo-SCT, we resequenced 35 genes in 113 adults at diagnosis, 49 of whom relapsed. Two hundred sixty-two mutations were detected in 102/113 (90%) patients. An increased risk of relapse was observed in patients with mutations in WT1 (P = .018), DNMT3A (P = .045), FLT3 ITD (P = .071), and TP53 (P = .06), whereas mutations in IDH1 were associated with a reduced risk of disease relapse (P = .018). In 29 patients, we additionally compared mutational profiles in bone marrow at diagnosis and relapse to study changes in clonal structure at relapse. In 13/29 patients, mutational profiles altered at relapse. In 9 patients, mutations present at relapse were not detected at diagnosis. In 15 patients, additional available pre–allo-SCT samples demonstrated that mutations identified posttransplant but not at diagnosis were detectable immediately prior to transplant in 2 of 15 patients. Taken together, these observations, if confirmed in larger studies, have the potential to inform the design of novel strategies to reduce posttransplant relapse highlighting the potential importance of post–allo-SCT interventions with a broad antitumor specificity in contrast to targeted therapies based on mutational profile at diagnosis.
- Published
- 2016
- Full Text
- View/download PDF
33. Genetically distinct leukemic stem cells in human CD34− acute myeloid leukemia are arrested at a hemopoietic precursor-like stage
- Author
-
Quek, Lynn, Otto, Georg W., Garnett, Catherine, Lhermitte, Ludovic, Karamitros, Dimitris, Stoilova, Bilyana, Lau, I-Jun, Doondeea, Jessica, Usukhbayar, Batchimeg, Kennedy, Alison, Metzner, Marlen, Goardon, Nicolas, Ivey, Adam, Allen, Christopher, Gale, Rosemary, Davies, Benjamin, Sternberg, Alexander, Killick, Sally, Hunter, Hannah, Cahalin, Paul, Price, Andrew, Carr, Andrew, Griffiths, Mike, Virgo, Paul, Mackinnon, Stephen, Grimwade, David, Freeman, Sylvie, Russell, Nigel, Craddock, Charles, Mead, Adam, Peniket, Andrew, Porcher, Catherine, and Vyas, Paresh
- Abstract
Our understanding of the perturbation of normal cellular differentiation hierarchies to create tumor-propagating stem cell populations is incomplete. In human acute myeloid leukemia (AML), current models suggest transformation creates leukemic stem cell (LSC) populations arrested at a progenitor-like stage expressing cell surface CD34. We show that in ∼25% of AML, with a distinct genetic mutation pattern where >98% of cells are CD34−, there are multiple, nonhierarchically arranged CD34+ and CD34− LSC populations. Within CD34− and CD34+ LSC–containing populations, LSC frequencies are similar; there are shared clonal structures and near-identical transcriptional signatures. CD34− LSCs have disordered global transcription profiles, but these profiles are enriched for transcriptional signatures of normal CD34− mature granulocyte–macrophage precursors, downstream of progenitors. But unlike mature precursors, LSCs express multiple normal stem cell transcriptional regulators previously implicated in LSC function. This suggests a new refined model of the relationship between LSCs and normal hemopoiesis in which the nature of genetic/epigenetic changes determines the disordered transcriptional program, resulting in LSC differentiation arrest at stages that are most like either progenitor or precursor stages of hemopoiesis.
- Published
- 2016
- Full Text
- View/download PDF
34. Clinical and biological implications of driver mutations in myelodysplastic syndromes
- Author
-
Papaemmanuil, Elli, Gerstung, Moritz, Malcovati, Luca, Tauro, Sudhir, Gundem, Gunes, Van Loo, Peter, Yoon, Chris J., Ellis, Peter, Wedge, David C., Pellagatti, Andrea, Shlien, Adam, Groves, Michael John, Forbes, Simon A., Raine, Keiran, Hinton, Jon, Mudie, Laura J., McLaren, Stuart, Hardy, Claire, Latimer, Calli, Della Porta, Matteo G., O’Meara, Sarah, Ambaglio, Ilaria, Galli, Anna, Butler, Adam P., Walldin, Gunilla, Teague, Jon W., Quek, Lynn, Sternberg, Alex, Gambacorti-Passerini, Carlo, Cross, Nicholas C. P., Green, Anthony R., Boultwood, Jacqueline, Vyas, Paresh, Hellstrom-Lindberg, Eva, Bowen, David, Cazzola, Mario, Stratton, Michael R., and Campbell, Peter J.
- Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of chronic hematological malignancies characterized by dysplasia, ineffective hematopoiesis and a variable risk of progression to acute myeloid leukemia. Sequencing of MDS genomes has identified mutations in genes implicated in RNA splicing, DNA modification, chromatin regulation, and cell signaling. We sequenced 111 genes across 738 patients with MDS or closely related neoplasms (including chronic myelomonocytic leukemia and MDS–myeloproliferative neoplasms) to explore the role of acquired mutations in MDS biology and clinical phenotype. Seventy-eight percent of patients had 1 or more oncogenic mutations. We identify complex patterns of pairwise association between genes, indicative of epistatic interactions involving components of the spliceosome machinery and epigenetic modifiers. Coupled with inferences on subclonal mutations, these data suggest a hypothesis of genetic “predestination,” in which early driver mutations, typically affecting genes involved in RNA splicing, dictate future trajectories of disease evolution with distinct clinical phenotypes. Driver mutations had equivalent prognostic significance, whether clonal or subclonal, and leukemia-free survival deteriorated steadily as numbers of driver mutations increased. Thus, analysis of oncogenic mutations in large, well-characterized cohorts of patients illustrates the interconnections between the cancer genome and disease biology, with considerable potential for clinical application.
- Published
- 2013
- Full Text
- View/download PDF
35. Clinical and biological implications of driver mutations in myelodysplastic syndromes
- Author
-
Papaemmanuil, Elli, Gerstung, Moritz, Malcovati, Luca, Tauro, Sudhir, Gundem, Gunes, Van Loo, Peter, Yoon, Chris J., Ellis, Peter, Wedge, David C., Pellagatti, Andrea, Shlien, Adam, Groves, Michael John, Forbes, Simon A., Raine, Keiran, Hinton, Jon, Mudie, Laura J., McLaren, Stuart, Hardy, Claire, Latimer, Calli, Della Porta, Matteo G., O'Meara, Sarah, Ambaglio, Ilaria, Galli, Anna, Butler, Adam P., Walldin, Gunilla, Teague, Jon W., Quek, Lynn, Sternberg, Alex, Gambacorti-Passerini, Carlo, Cross, Nicholas C.P., Green, Anthony R., Boultwood, Jacqueline, Vyas, Paresh, Hellstrom-Lindberg, Eva, Bowen, David, Cazzola, Mario, Stratton, Michael R., and Campbell, Peter J.
- Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of chronic hematological malignancies characterized by dysplasia, ineffective hematopoiesis and a variable risk of progression to acute myeloid leukemia. Sequencing of MDS genomes has identified mutations in genes implicated in RNA splicing, DNA modification, chromatin regulation, and cell signaling. We sequenced 111 genes across 738 patients with MDS or closely related neoplasms (including chronic myelomonocytic leukemia and MDS–myeloproliferative neoplasms) to explore the role of acquired mutations in MDS biology and clinical phenotype. Seventy-eight percent of patients had 1 or more oncogenic mutations. We identify complex patterns of pairwise association between genes, indicative of epistatic interactions involving components of the spliceosome machinery and epigenetic modifiers. Coupled with inferences on subclonal mutations, these data suggest a hypothesis of genetic “predestination,” in which early driver mutations, typically affecting genes involved in RNA splicing, dictate future trajectories of disease evolution with distinct clinical phenotypes. Driver mutations had equivalent prognostic significance, whether clonal or subclonal, and leukemia-free survival deteriorated steadily as numbers of driver mutations increased. Thus, analysis of oncogenic mutations in large, well-characterized cohorts of patients illustrates the interconnections between the cancer genome and disease biology, with considerable potential for clinical application.
- Published
- 2013
- Full Text
- View/download PDF
36. Acute myeloid leukemia creates an arginase-dependent immunosuppressive microenvironment
- Author
-
Mussai, Francis, De Santo, Carmela, Abu-Dayyeh, Issa, Booth, Sarah, Quek, Lynn, McEwen-Smith, Rosanna M., Qureshi, Amrana, Dazzi, Francesco, Vyas, Paresh, and Cerundolo, Vincenzo
- Abstract
Acute myeloid leukemia (AML) is the most common acute leukemia in adults and the second most common frequent leukemia of childhood. Patients may present with lymphopenia or pancytopenia at diagnosis. We investigated the mechanisms by which AML causes pancytopenia and suppresses patients’ immune response. This study identified for the first time that AML blasts alter the immune microenvironment through enhanced arginine metabolism. Arginase II is expressed and released from AML blasts and is present at high concentrations in the plasma of patients with AML, resulting in suppression of T-cell proliferation. We extended these results by demonstrating an arginase-dependent ability of AML blasts to polarize surrounding monocytes into a suppressive M2-like phenotype in vitro and in engrafted nonobese diabetic–severe combined immunodeficiency mice. In addition, AML blasts can suppress the proliferation and differentiation of murine granulocyte-monocyte progenitors and human CD34+ progenitors. Finally, the study showed that the immunosuppressive activity of AML blasts can be modulated through small-molecule inhibitors of arginase and inducible nitric oxide synthase, suggesting a novel therapeutic target in AML. The results strongly support the hypothesis that AML creates an immunosuppressive microenvironment that contributes to the pancytopenia observed at diagnosis.
- Published
- 2013
- Full Text
- View/download PDF
37. Acute myeloid leukemia creates an arginase-dependent immunosuppressive microenvironment
- Author
-
Mussai, Francis, De Santo, Carmela, Abu-Dayyeh, Issa, Booth, Sarah, Quek, Lynn, McEwen-Smith, Rosanna M., Qureshi, Amrana, Dazzi, Francesco, Vyas, Paresh, and Cerundolo, Vincenzo
- Abstract
Acute myeloid leukemia (AML) is the most common acute leukemia in adults and the second most common frequent leukemia of childhood. Patients may present with lymphopenia or pancytopenia at diagnosis. We investigated the mechanisms by which AML causes pancytopenia and suppresses patients' immune response. This study identified for the first time that AML blasts alter the immune microenvironment through enhanced arginine metabolism. Arginase II is expressed and released from AML blasts and is present at high concentrations in the plasma of patients with AML, resulting in suppression of T-cell proliferation. We extended these results by demonstrating an arginase-dependent ability of AML blasts to polarize surrounding monocytes into a suppressive M2-like phenotype in vitro and in engrafted nonobese diabetic–severe combined immunodeficiency mice. In addition, AML blasts can suppress the proliferation and differentiation of murine granulocyte-monocyte progenitors and human CD34+progenitors. Finally, the study showed that the immunosuppressive activity of AML blasts can be modulated through small-molecule inhibitors of arginase and inducible nitric oxide synthase, suggesting a novel therapeutic target in AML. The results strongly support the hypothesis that AML creates an immunosuppressive microenvironment that contributes to the pancytopenia observed at diagnosis.
- Published
- 2013
- Full Text
- View/download PDF
38. Genetic and Pharmacological Analyses of Involvement of Src-family, Syk and Btk Tyrosine Kinases in Platelet Shape Change
- Author
-
Bauer, Markus, Maschberger, Petra, Quek, Lynn, Briddon, Stephen J., Dash, Debabrata, Weiss, Michael, Watson, Steve P., and Siess, Wolfgang
- Published
- 2001
- Full Text
- View/download PDF
39. Fyn and Lyn phosphorylate the Fc receptor ? chain downstream of glycoprotein VI in murine platelets, and Lyn regulates a novel feedback pathway
- Author
-
Quek, Lynn S., Pasquet, Jean-Max, Hers, Ingeborg, Cornall, Richard, Knight, Graham, Barnes, Michael, Hibbs, Margaret L., Dunn, Ashley R., Lowell, Clifford A., and Watson, Steve P.
- Abstract
Activation of platelets by collagen is mediated by the complex glycoprotein VI (GPVI)/Fc receptor ? (FcR? chain). In the current study, the role of 2 Src family kinases, Fyn and Lyn, in GPVI signaling has been examined using murine platelets deficient in one or both kinases. In the fyn-/-platelets, tyrosine phosphorylation of FcR? chain, phopholipase C (PLC) activity, aggregation, and secretion are reduced, though the time of onset of response is unchanged. In the lyn-/-platelets, there is a delay of up to 30 seconds in the onset of tyrosine phosphorylation and functional responses, followed by recovery of phosphorylation and potentiation of aggregation and a-granule secretion. Tyrosine phosphorylation and aggregation in response to stimulation by collagen-related peptide is further attenuated and delayed in fyn-/-lyn-/-double-mutant platelets, and potentiation is not seen. This study provides the first genetic evidence that Fyn and Lyn mediate FcR immune receptor tyrosine-based activation motif phosphorylation and PLC?2 activation after the ligation of GPVI. Lyn plays an additional role in inhibiting platelet activation through an uncharacterized inhibitory pathway.
- Published
- 2000
- Full Text
- View/download PDF
40. Fyn and Lyn phosphorylate the Fc receptor γ chain downstream of glycoprotein VI in murine platelets, and Lyn regulates a novel feedback pathway
- Author
-
Quek, Lynn S., Pasquet, Jean-Max, Hers, Ingeborg, Cornall, Richard, Knight, Graham, Barnes, Michael, Hibbs, Margaret L., Dunn, Ashley R., Lowell, Clifford A., and Watson, Steve P.
- Abstract
Activation of platelets by collagen is mediated by the complex glycoprotein VI (GPVI)/Fc receptor γ (FcRγ chain). In the current study, the role of 2 Src family kinases, Fyn and Lyn, in GPVI signaling has been examined using murine platelets deficient in one or both kinases. In the fyn−/−platelets, tyrosine phosphorylation of FcRγ chain, phopholipase C (PLC) activity, aggregation, and secretion are reduced, though the time of onset of response is unchanged. In the lyn−/−platelets, there is a delay of up to 30 seconds in the onset of tyrosine phosphorylation and functional responses, followed by recovery of phosphorylation and potentiation of aggregation and α-granule secretion. Tyrosine phosphorylation and aggregation in response to stimulation by collagen-related peptide is further attenuated and delayed in fyn−/−lyn−/−double-mutant platelets, and potentiation is not seen. This study provides the first genetic evidence that Fyn and Lyn mediate FcR immune receptor tyrosine-based activation motif phosphorylation and PLCγ2 activation after the ligation of GPVI. Lyn plays an additional role in inhibiting platelet activation through an uncharacterized inhibitory pathway.
- Published
- 2000
- Full Text
- View/download PDF
41. Thrombopoietin potentiates collagen receptor signaling in platelets through a phosphatidylinositol 3-kinase–dependent pathway
- Author
-
Pasquet, Jean-max, Gross, Barbara S., Gratacap, Marie-Pierre, Quek, Lynn, Pasquet, Sophie, Payrastre, Bernard, van Willigen, Gijsbert, Mountford, Joanne C., and Watson, Steve P.
- Abstract
Collagen activates platelets through a tyrosine kinase-dependent pathway, involving phospholipase C?2. Functional responses such as aggregation and secretion induced by collagen are potentiated by preincubation with thrombopoietin (TPO). In this study, we show that collagen and thrombopoietin activate the phosphatidylinositol 3-kinase (PI 3-kinase) pathway and that this contributes to their respective actions. The structurally distinct inhibitors of PI 3-kinase, wortmannin, and LY294002, completely inhibit formation of phosphatidylinositol 3,4,5-trisphosphate by collagen. This leads to a substantial reduction in the formation of inositol phosphates and phosphatidic acid, 2 indices of PLC activity, and the consequent inhibition of intracellular Ca++[Ca++]i, aggregation and secretion. Potentiation of the collagen response by TPO is prevented in the presence of wortmannin and LY294002. However, when the 2 PI 3-kinase inhibitors are given after the addition of TPO but before the collagen, recovery of potentiation is observed. This suggests that potentiation is mediated through activation of PI 3-kinase. TPO stimulates aggregation of platelets from a low percentage of donors and this is also blocked by wortmannin. These results suggest that the PI 3-kinase pathway plays an important role in signaling by collagen and in the priming action of TPO.
- Published
- 2000
- Full Text
- View/download PDF
42. Thrombopoietin potentiates collagen receptor signaling in platelets through a phosphatidylinositol 3-kinase–dependent pathway
- Author
-
Pasquet, Jean-max, Gross, Barbara S., Gratacap, Marie-Pierre, Quek, Lynn, Pasquet, Sophie, Payrastre, Bernard, van Willigen, Gijsbert, Mountford, Joanne C., and Watson, Steve P.
- Abstract
Collagen activates platelets through a tyrosine kinase-dependent pathway, involving phospholipase Cγ2. Functional responses such as aggregation and secretion induced by collagen are potentiated by preincubation with thrombopoietin (TPO). In this study, we show that collagen and thrombopoietin activate the phosphatidylinositol 3-kinase (PI 3-kinase) pathway and that this contributes to their respective actions. The structurally distinct inhibitors of PI 3-kinase, wortmannin, and LY294002, completely inhibit formation of phosphatidylinositol 3,4,5-trisphosphate by collagen. This leads to a substantial reduction in the formation of inositol phosphates and phosphatidic acid, 2 indices of PLC activity, and the consequent inhibition of intracellular Ca++[Ca++]i, aggregation and secretion. Potentiation of the collagen response by TPO is prevented in the presence of wortmannin and LY294002. However, when the 2 PI 3-kinase inhibitors are given after the addition of TPO but before the collagen, recovery of potentiation is observed. This suggests that potentiation is mediated through activation of PI 3-kinase. TPO stimulates aggregation of platelets from a low percentage of donors and this is also blocked by wortmannin. These results suggest that the PI 3-kinase pathway plays an important role in signaling by collagen and in the priming action of TPO.
- Published
- 2000
- Full Text
- View/download PDF
43. Repressed Chromatin Drives Leukaemogenesis in Mutant IDH2 Acute Myeloid Leukaemia Via Inhibition of Granulocyte Differentiation and Cell Cycle Progression
- Author
-
Silveira, Douglas RA, Chatzikyriakou, Prodromos, Yavorska, Olena, Mackie, Sarah, Hulks, Roan, Siejka-Zielinska, Paulina, Corby, Anna, Smith, Alastair, Brown, Benjamin, Metzner, Marlen, Milne, Thomas A., Penard-Lacronique, Virginie, Song, Chunxiao, Hasan, Maroof, Thakurta, Anjan, Vyas, Paresh, Kriaucionis, Skirmantas, and Quek, Lynn
- Abstract
Differentiation arrest in acute myeloid leukaemia (AML) results in accumulation of leukaemic progenitors (L-Prog) and bone marrow failure. Mutant isocitrate dehydrogenase enzyme produces d-2-hydroxyglutarate (2HG), which inhibits α-ketoglutarate-dependent dioxygenases, including Jumonji histone demethylases (JKDM) and TET2, but how this causes AML is unclear. Inhibitors of mutant IDH enzyme (mIDHi) restore differentiation in IDH-mutant (mIDH) AML (Amatangelo et al., 2018). Here, we studied transcriptional networks involved using single-cell (SC) gene expression (GEX) and transcription factor (TF) motif accessibility in primary AML treated with the mIDH2 inhibitor enasidenib (ENA) and found that ENA activates cell cycle (CC) and pro-differentiation programmes through increased promoter accessibility of granulocyte-monocyte (GM)-TF targets.
- Published
- 2021
- Full Text
- View/download PDF
44. Tumor Associated Macrophages Promoted Fatal Patient Derived Acute Promyelocytic Leukemia In Vivo
- Author
-
Weinhäuser, Isabel, Pereira-Martins, Diego A, Hilberink, Jacobien R, Almeida, Luciana Yamamoto, Silveira, Douglas RA, Quek, Lynn, Hogeling, Shanna M, Mota, Jose Mauricio, Ammatuna, Emanuele, Lucena-Araujo, Antonio R., Huls, Gerwin A., Schuringa, Jan Jacob, and Rego, Eduardo M
- Abstract
With immune therapies on the rise, an in-depth understanding of the immunological changes in leukemic bone marrow (BM) niches becomes indispensable. Being an crucial part of the tumor microenvironment (TME) in solid tumours, tumour-associated macrophages are often associated with poor prognosis (Bruni et al. 2020). Yet, in acute myeloid leukaemia (AML) the role of macrophages has not been thoroughly studied. The expression of the M2-markers CD163 and CD206 in the AML BM cell population predicted poor clinical outcome. We identified that this expression emerges from a more mature (CD45 midSSC highHLA-DR +CD14 +CD16 +/-) myeloid cell population (hereafter called AML-associated macrophages - AAM) and not from the leukemic blasts. By employing flow cytometry analysis (FACS) we noted a decrease in the expression of the M1-marker (CD80) and an increase of the M2-markers CD163/CD206on AAM (n=70) compared to healthy donors (HD, n=10). Unsupervised clustering based on the CD163/CD206 levels detected on AAM generated 4 distinct clusters, whereby patients within the CD163 low/CD206 lowcluster displayed better overall survival than the other clusters.
- Published
- 2021
- Full Text
- View/download PDF
45. Tumor Associated Macrophages Promoted Fatal Patient Derived Acute Promyelocytic Leukemia In Vivo
- Author
-
Weinhäuser, Isabel, Pereira-Martins, Diego A, Hilberink, Jacobien R, Almeida, Luciana Yamamoto, Silveira, Douglas RA, Quek, Lynn, Hogeling, Shanna M, Mota, Jose Mauricio, Ammatuna, Emanuele, Lucena-Araujo, Antonio R., Huls, Gerwin A., Schuringa, Jan Jacob, and Rego, Eduardo M
- Abstract
Silveira: BMS/Celgene: Research Funding; Servier/Agios: Research Funding; Abbvie: Speakers Bureau; Astellas: Speakers Bureau. Quek: BMS/Celgene: Research Funding; Servier/Agios: Research Funding. Mota: Janssen: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Astellas: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Technopharma: Speakers Bureau; Bristol Myer Squibb: Speakers Bureau; Bayer: Speakers Bureau; Pfizer: Speakers Bureau; AstraZeneca: Speakers Bureau; Astellas: Speakers Bureau; Ipsen: Speakers Bureau; Amgen: Speakers Bureau.
- Published
- 2021
- Full Text
- View/download PDF
46. Repressed Chromatin Drives Leukaemogenesis in Mutant IDH2 Acute Myeloid Leukaemia Via Inhibition of Granulocyte Differentiation and Cell Cycle Progression
- Author
-
Silveira, Douglas RA, Chatzikyriakou, Prodromos, Yavorska, Olena, Mackie, Sarah, Hulks, Roan, Siejka-Zielinska, Paulina, Corby, Anna, Smith, Alastair, Brown, Benjamin, Metzner, Marlen, Milne, Thomas A., Penard-Lacronique, Virginie, Song, Chunxiao, Hasan, Maroof, Thakurta, Anjan, Vyas, Paresh, Kriaucionis, Skirmantas, and Quek, Lynn
- Abstract
Silveira: Astellas: Speakers Bureau; Abbvie: Speakers Bureau; Servier/Agios: Research Funding; BMS/Celgene: Research Funding. Hasan: Bristol Myers Squibb: Current Employment. Thakurta: Bristol Myers Squibb: Current Employment, Current equity holder in publicly-traded company, Patents & Royalties. Vyas: Gilead: Honoraria; Astellas: Consultancy, Honoraria; AbbVie: Consultancy, Honoraria; Takeda: Honoraria; Bristol Myers Squibb: Consultancy, Honoraria, Research Funding; Janssen: Honoraria; Daiichi Sankyo: Honoraria; Jazz: Honoraria; Pfizer: Honoraria; Novartis: Honoraria. Quek: BMS/Celgene: Research Funding; Servier/Agios: Research Funding.
- Published
- 2021
- Full Text
- View/download PDF
47. Regulation and Function of WASp in Platelets by the Collagen Receptor, Glycoprotein VI
- Author
-
Gross, Barbara S., Wilde, Jonathan I., Quek, Lynn, Chapel, Helen, Nelson, David L., and Watson, Steve P.
- Abstract
Wiskott Aldrich syndrome (WAS) is an X-linked recessive disorder associated with abnormalities in platelets and lymphocytes giving rise to thrombocytopenia and immunodeficiency. WAS is caused by a mutation in the gene encoding the cytoskeletal protein (WASp). Despite its importance, the role of WASp in platelet function is not established. WASp was recently shown to undergo tyrosine phosphorylation in platelets after activation by collagen, suggesting that it may play a selective role in activation by the adhesion molecule. In the present study, we show that WASp is heavily tyrosine phosphorylated by a collagen-related peptide (CRP) that binds to the collagen receptor glycoprotein (GP) VI, but not to the integrin ?2ß1. Tyrosine phosphorylation of WASp was blocked by Src family kinase inhibitors and reduced by treatment with wortmannin and in patients with X-linked agammaglobulinemia (XLA), a condition caused by a lack of functional expression of Btk. This indicates that Src kinases, phosphatidylinositol 3-kinase (PI 3-kinase), and Btk all contribute to the regulation of tyrosine phosphorylation of WASp. The functional importance of WASp was investigated in 2 WAS brothers who show no detectable expression of WASp. Platelet aggregation and secretion from dense granules induced by CRP and thrombin was slightly enhanced in the WAS platelets relative to controls. Furthermore, there was no apparent difference in morphology in WAS platelets after stimulation by these agonists. These observations suggest that WASp does not play a critical role in intracellular signaling downstream of tyrosine kinase-linked and G protein-coupled receptors in platelets.
- Published
- 1999
- Full Text
- View/download PDF
48. Regulation and Function of WASp in Platelets by the Collagen Receptor, Glycoprotein VI
- Author
-
Gross, Barbara S., Wilde, Jonathan I., Quek, Lynn, Chapel, Helen, Nelson, David L., and Watson, Steve P.
- Abstract
Wiskott Aldrich syndrome (WAS) is an X-linked recessive disorder associated with abnormalities in platelets and lymphocytes giving rise to thrombocytopenia and immunodeficiency. WAS is caused by a mutation in the gene encoding the cytoskeletal protein (WASp). Despite its importance, the role of WASp in platelet function is not established. WASp was recently shown to undergo tyrosine phosphorylation in platelets after activation by collagen, suggesting that it may play a selective role in activation by the adhesion molecule. In the present study, we show that WASp is heavily tyrosine phosphorylated by a collagen-related peptide (CRP) that binds to the collagen receptor glycoprotein (GP) VI, but not to the integrin α2β1. Tyrosine phosphorylation of WASp was blocked by Src family kinase inhibitors and reduced by treatment with wortmannin and in patients with X-linked agammaglobulinemia (XLA), a condition caused by a lack of functional expression of Btk. This indicates that Src kinases, phosphatidylinositol 3-kinase (PI 3-kinase), and Btk all contribute to the regulation of tyrosine phosphorylation of WASp. The functional importance of WASp was investigated in 2 WAS brothers who show no detectable expression of WASp. Platelet aggregation and secretion from dense granules induced by CRP and thrombin was slightly enhanced in the WAS platelets relative to controls. Furthermore, there was no apparent difference in morphology in WAS platelets after stimulation by these agonists. These observations suggest that WASp does not play a critical role in intracellular signaling downstream of tyrosine kinase-linked and G protein-coupled receptors in platelets.
- Published
- 1999
- Full Text
- View/download PDF
49. LAT Is Required for Tyrosine Phosphorylation of Phospholipase C?2 and Platelet Activation by the Collagen Receptor GPVI
- Author
-
Pasquet, Jean-Max, Gross, Barbara, Quek, Lynn, Asazuma, Naoki, Zhang, Weiguo, Sommers, Connie L., Schweighoffer, Edina, Tybulewicz, Victor, Judd, Barbara, Lee, Jong Ran, Koretzky, Gary, Love, Paul E., Samelson, Lawrence E., and Watson, Steve P.
- Abstract
ABSTRACTIn the present study, we have addressed the role of the linker for activation of T cells (LAT) in the regulation of phospholipase C?2 (PLC?2) by the platelet collagen receptor glycoprotein VI (GPVI). LAT is tyrosine phosphorylated in human platelets heavily in response to collagen, collagen-related peptide (CRP), and Fc?RIIA cross-linking but only weakly in response to the G-protein-receptor-coupled agonist thrombin. LAT tyrosine phosphorylation is abolished in CRP-stimulated Syk-deficient mouse platelets, whereas it is not altered in SLP-76-deficient mice or Btk-deficient X-linked agammaglobulinemia (XLA) human platelets. Using mice engineered to lack the adapter LAT, we showed that tyrosine phosphorylation of Syk and Btk in response to CRP was maintained in LAT-deficient platelets whereas phosphorylation of SLP-76 was slightly impaired. In contrast, tyrosine phosphorylation of PLC?2 was substantially reduced in LAT-deficient platelets but was not completely inhibited. The reduction in phosphorylation of PLC?2 was associated with marked inhibition of formation of phosphatidic acid, a metabolite of 1,2-diacylglycerol, phosphorylation of pleckstrin, a substrate of protein kinase C, and expression of P-selectin in response to CRP, whereas these parameters were not altered in response to thrombin. Activation of the fibrinogen receptor integrin aIIbß3in response to CRP was also reduced in LAT-deficient platelets but was not completely inhibited. These results demonstrate that LAT tyrosine phosphorylation occurs downstream of Syk and is independent of the adapter SLP-76, and they establish a major role for LAT in the phosphorylation and activation of PLC?2, leading to downstream responses such as a-granule secretion and activation of integrin aIIbß3. The results further demonstrate that the major pathway of tyrosine phosphorylation of SLP-76 is independent of LAT and that there is a minor, LAT-independent pathway of tyrosine phosphorylation of PLC?2. We propose a model in which LAT and SLP-76 are required for PLC?2 phosphorylation but are regulated through independent pathways downstream of Syk.
- Published
- 1999
- Full Text
- View/download PDF
50. LAT Is Required for Tyrosine Phosphorylation of Phospholipase Cγ2 and Platelet Activation by the Collagen Receptor GPVI
- Author
-
Pasquet, Jean-Max, Gross, Barbara, Quek, Lynn, Asazuma, Naoki, Zhang, Weiguo, Sommers, Connie L., Schweighoffer, Edina, Tybulewicz, Victor, Judd, Barbara, Lee, Jong Ran, Koretzky, Gary, Love, Paul E., Samelson, Lawrence E., and Watson, Steve P.
- Abstract
In the present study, we have addressed the role of the linker for activation of T cells (LAT) in the regulation of phospholipase Cγ2 (PLCγ2) by the platelet collagen receptor glycoprotein VI (GPVI). LAT is tyrosine phosphorylated in human platelets heavily in response to collagen, collagen-related peptide (CRP), and FcγRIIA cross-linking but only weakly in response to the G-protein-receptor-coupled agonist thrombin. LAT tyrosine phosphorylation is abolished in CRP-stimulated Syk-deficient mouse platelets, whereas it is not altered in SLP-76-deficient mice or Btk-deficient X-linked agammaglobulinemia (XLA) human platelets. Using mice engineered to lack the adapter LAT, we showed that tyrosine phosphorylation of Syk and Btk in response to CRP was maintained in LAT-deficient platelets whereas phosphorylation of SLP-76 was slightly impaired. In contrast, tyrosine phosphorylation of PLCγ2 was substantially reduced in LAT-deficient platelets but was not completely inhibited. The reduction in phosphorylation of PLCγ2 was associated with marked inhibition of formation of phosphatidic acid, a metabolite of 1,2-diacylglycerol, phosphorylation of pleckstrin, a substrate of protein kinase C, and expression of P-selectin in response to CRP, whereas these parameters were not altered in response to thrombin. Activation of the fibrinogen receptor integrin αIIbβ3in response to CRP was also reduced in LAT-deficient platelets but was not completely inhibited. These results demonstrate that LAT tyrosine phosphorylation occurs downstream of Syk and is independent of the adapter SLP-76, and they establish a major role for LAT in the phosphorylation and activation of PLCγ2, leading to downstream responses such as α-granule secretion and activation of integrin αIIbβ3. The results further demonstrate that the major pathway of tyrosine phosphorylation of SLP-76 is independent of LAT and that there is a minor, LAT-independent pathway of tyrosine phosphorylation of PLCγ2. We propose a model in which LAT and SLP-76 are required for PLCγ2 phosphorylation but are regulated through independent pathways downstream of Syk.
- Published
- 1999
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.