333 results on '"Adolfo A. Ferrando"'
Search Results
2. Phosphoproteomic profiling of T cell acute lymphoblastic leukemia reveals targetable kinases and combination treatment strategies
- Author
-
Valentina Cordo’, Mariska T. Meijer, Rico Hagelaar, Richard R. de Goeij-de Haas, Vera M. Poort, Alex A. Henneman, Sander R. Piersma, Thang V. Pham, Koichi Oshima, Adolfo A. Ferrando, Guido J. R. Zaman, Connie R. Jimenez, and Jules P. P. Meijerink
- Subjects
Science - Abstract
No targeted therapy has been approved yet for the treatment of T cell acute lymphoblastic leukemia. Here the authors show that unbiased phosphoproteomic profiling can identify targetable kinases and guide the design of personalized combination treatments using kinase inhibitors.
- Published
- 2022
- Full Text
- View/download PDF
3. Epigenetic reversal of hematopoietic stem cell aging in Phf6-knockout mice
- Author
-
Agnieszka A. Wendorff, S. Aidan Quinn, Silvia Alvarez, Jessie A. Brown, Mayukh Biswas, Thomas Gunning, Teresa Palomero, and Adolfo A. Ferrando
- Subjects
Aging ,Neuroscience (miscellaneous) ,Geriatrics and Gerontology - Published
- 2022
- Full Text
- View/download PDF
4. Data from Intracellular Cholesterol Pools Regulate Oncogenic Signaling and Epigenetic Circuitries in Early T-cell Precursor Acute Lymphoblastic Leukemia
- Author
-
Adolfo A. Ferrando, Rajesh K. Soni, Maria Luisa Sulis, Selina Luger, Peter H. Wiernik, Mark R. Litzow, Jacob M. Rowe, Martin S. Tallman, Elisabeth M. Paietta, Adam L. Mackey, Hannah I. Miller, Pablo Perez-Duran, Francesca Gianni, Robert Albero, and Marissa Rashkovan
- Abstract
Early T-cell acute lymphoblastic leukemia (ETP-ALL) is an aggressive hematologic malignancy associated with early relapse and poor prognosis that is genetically, immunophenotypically, and transcriptionally distinct from more mature T-cell acute lymphoblastic leukemia (T-ALL) tumors. Here, we leveraged global metabolomic and transcriptomic profiling of primary ETP- and T-ALL leukemia samples to identify specific metabolic circuitries differentially active in this high-risk leukemia group. ETP-ALLs showed increased biosynthesis of phospholipids and sphingolipids and were specifically sensitive to inhibition of 3-hydroxy-3-methylglutaryl-CoA reductase, the rate-limiting enzyme in the mevalonate pathway. Mechanistically, inhibition of cholesterol synthesis inhibited oncogenic AKT1 signaling and suppressed MYC expression via loss of chromatin accessibility at a leukemia stem cell–specific long-range MYC enhancer. In all, these results identify the mevalonate pathway as a druggable novel vulnerability in high-risk ETP-ALL cells and uncover an unanticipated critical role for cholesterol biosynthesis in signal transduction and epigenetic circuitries driving leukemia cell growth and survival.Significance:Overtly distinct cell metabolic pathways operate in ETP- and T-ALL pointing to specific metabolic vulnerabilities. Inhibition of mevalonate biosynthesis selectively blocks oncogenic AKT–MYC signaling in ETP-ALL and suppresses leukemia cell growth. Ultimately, these results will inform the development of novel tailored and more effective treatments for patients with high-risk ETP-ALL.This article is highlighted in the In This Issue feature, p. 587
- Published
- 2023
- Full Text
- View/download PDF
5. Data from Pharmacologic Inhibition of NT5C2 Reverses Genetic and Nongenetic Drivers of 6-MP Resistance in Acute Lymphoblastic Leukemia
- Author
-
Adolfo A. Ferrando, Teresa Palomero, Brent R. Stockwell, Liang Tong, Mignon L. Loh, Julie M. Gastier-Foster, Cindy Ma, Hannah I. Miller, Robert Albero, Wen-Hsuan W. Lin, Anouchka P. Laurent, Farhad Forouhar, Arie Zask, Chelsea L. Dieck, and Clara Reglero
- Abstract
Low-intensity maintenance therapy with 6-mercaptopurine (6-MP) limits the occurrence of acute lymphoblastic leukemia (ALL) relapse and is central to the success of multiagent chemotherapy protocols. Activating mutations in the 5′-nucleotidase cytosolic II (NT5C2) gene drive resistance to 6-MP in over 35% of early relapse ALL cases. Here we identify CRCD2 as a first-in-class small-molecule NT5C2 nucleotidase inhibitor broadly active against leukemias bearing highly prevalent relapse-associated mutant forms of NT5C2 in vitro and in vivo. Importantly, CRCD2 treatment also enhanced the cytotoxic activity of 6-MP in NT5C2 wild-type leukemias, leading to the identification of NT5C2 Ser502 phosphorylation as a novel NT5C2-mediated mechanism of 6-MP resistance in this disease. These results uncover an unanticipated role of nongenetic NT5C2 activation as a driver of 6-MP resistance in ALL and demonstrate the potential of NT5C2 inhibitor therapy for enhancing the efficacy of thiopurine maintenance therapy and overcoming resistance at relapse.Significance:Relapse-associated NT5C2 mutations directly contribute to relapse in ALL by driving resistance to chemotherapy with 6-MP. Pharmacologic inhibition of NT5C2 with CRCD2, a first-in-class nucleotidase inhibitor, enhances the cytotoxic effects of 6-MP and effectively reverses thiopurine resistance mediated by genetic and nongenetic mechanisms of NT5C2 activation in ALL.This article is highlighted in the In This Issue feature, p. 2483
- Published
- 2023
- Full Text
- View/download PDF
6. Data from Targeting S100A9–ALDH1A1–Retinoic Acid Signaling to Suppress Brain Relapse in EGFR-Mutant Lung Cancer
- Author
-
Swarnali Acharyya, Joan Massague, Mark G. Kris, Catherine A. Shu, Anjali Saqi, Paul K. Paik, Helena A. Yu, Peter Canoll, Cathy Mendelsohn, Adolfo A. Ferrando, Elisa de Stanchina, Katia Manova-Todorova, Angeliki Mela, Galina G. Lagos, Hanna Scholze, Timothy James Zhong, Ahmad Rushdi Shakri, S. Aidan Quinn, Courtney Coker, Wanchao Ma, Yifan Tai, Seoyoung Han, and Anup Kumar Biswas
- Abstract
The epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) osimertinib has significantly prolonged progression-free survival (PFS) in patients with EGFR-mutant lung cancer, including those with brain metastases. However, despite striking initial responses, osimertinib-treated patients eventually develop lethal metastatic relapse, often to the brain. Although osimertinib-refractory brain relapse is a major clinical challenge, its underlying mechanisms remain poorly understood. Using metastatic models of EGFR-mutant lung cancer, we show that cancer cells expressing high intracellular S100A9 escape osimertinib and initiate brain relapses. Mechanistically, S100A9 upregulates ALDH1A1 expression and activates the retinoic acid (RA) signaling pathway in osimertinib-refractory cancer cells. We demonstrate that the genetic repression of S100A9, ALDH1A1, or RA receptors (RAR) in cancer cells, or treatment with a pan-RAR antagonist, dramatically reduces brain metastasis. Importantly, S100A9 expression in cancer cells correlates with poor PFS in osimertinib-treated patients. Our study, therefore, identifies a novel, therapeutically targetable S100A9–ALDH1A1–RA axis that drives brain relapse.Significance:Treatment with the EGFR TKI osimertinib prolongs the survival of patients with EGFR-mutant lung cancer; however, patients develop metastatic relapses, often to the brain. We identified a novel intracellular S100A9–ALDH1A1–RA signaling pathway that drives lethal brain relapse and can be targeted by pan-RAR antagonists to prevent cancer progression and prolong patient survival.This article is highlighted in the In This Issue feature, p. 873
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figure from Pharmacologic Inhibition of NT5C2 Reverses Genetic and Nongenetic Drivers of 6-MP Resistance in Acute Lymphoblastic Leukemia
- Author
-
Adolfo A. Ferrando, Teresa Palomero, Brent R. Stockwell, Liang Tong, Mignon L. Loh, Julie M. Gastier-Foster, Cindy Ma, Hannah I. Miller, Robert Albero, Wen-Hsuan W. Lin, Anouchka P. Laurent, Farhad Forouhar, Arie Zask, Chelsea L. Dieck, and Clara Reglero
- Abstract
Supplementary Figure from Pharmacologic Inhibition of NT5C2 Reverses Genetic and Nongenetic Drivers of 6-MP Resistance in Acute Lymphoblastic Leukemia
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure from Targeting S100A9–ALDH1A1–Retinoic Acid Signaling to Suppress Brain Relapse in EGFR-Mutant Lung Cancer
- Author
-
Swarnali Acharyya, Joan Massague, Mark G. Kris, Catherine A. Shu, Anjali Saqi, Paul K. Paik, Helena A. Yu, Peter Canoll, Cathy Mendelsohn, Adolfo A. Ferrando, Elisa de Stanchina, Katia Manova-Todorova, Angeliki Mela, Galina G. Lagos, Hanna Scholze, Timothy James Zhong, Ahmad Rushdi Shakri, S. Aidan Quinn, Courtney Coker, Wanchao Ma, Yifan Tai, Seoyoung Han, and Anup Kumar Biswas
- Abstract
Supplementary Figure from Targeting S100A9–ALDH1A1–Retinoic Acid Signaling to Suppress Brain Relapse in EGFR-Mutant Lung Cancer
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Table from Pharmacologic Inhibition of NT5C2 Reverses Genetic and Nongenetic Drivers of 6-MP Resistance in Acute Lymphoblastic Leukemia
- Author
-
Adolfo A. Ferrando, Teresa Palomero, Brent R. Stockwell, Liang Tong, Mignon L. Loh, Julie M. Gastier-Foster, Cindy Ma, Hannah I. Miller, Robert Albero, Wen-Hsuan W. Lin, Anouchka P. Laurent, Farhad Forouhar, Arie Zask, Chelsea L. Dieck, and Clara Reglero
- Abstract
Supplementary Table from Pharmacologic Inhibition of NT5C2 Reverses Genetic and Nongenetic Drivers of 6-MP Resistance in Acute Lymphoblastic Leukemia
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Data from Intracellular Cholesterol Pools Regulate Oncogenic Signaling and Epigenetic Circuitries in Early T-cell Precursor Acute Lymphoblastic Leukemia
- Author
-
Adolfo A. Ferrando, Rajesh K. Soni, Maria Luisa Sulis, Selina Luger, Peter H. Wiernik, Mark R. Litzow, Jacob M. Rowe, Martin S. Tallman, Elisabeth M. Paietta, Adam L. Mackey, Hannah I. Miller, Pablo Perez-Duran, Francesca Gianni, Robert Albero, and Marissa Rashkovan
- Abstract
Supplementary Data from Intracellular Cholesterol Pools Regulate Oncogenic Signaling and Epigenetic Circuitries in Early T-cell Precursor Acute Lymphoblastic Leukemia
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Table S2 from PRC2 Inhibitors Overcome Glucocorticoid Resistance Driven by NSD2 Mutation in Pediatric Acute Lymphoblastic Leukemia
- Author
-
Jonathan D. Licht, Richard B. Lock, Matthew D. Hall, Adolfo A. Ferrando, Richard L. Bennett, Alberto Riva, Min Shen, Christine M. Will, Jon A. Oyer, Marta Kulis, Alok Swaroop, Catalina Troche, Heidi L. Casellas Román, Duohui Jing, Jacob S. Roth, Daphne Dupéré-Richér, Crissandra Piper, Jonathan H. Shrimp, Julia Hlavka-Zhang, and Jianping Li
- Abstract
ALL patients with NSD2 mutations
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figure Legend from Preclinical Analysis of the γ-Secretase Inhibitor PF-03084014 in Combination with Glucocorticoids in T-cell Acute Lymphoblastic Leukemia
- Author
-
Adolfo A. Ferrando, Carlos Cordon-Cardo, Sophia Randolph, John Jakubczak, Jacob M. Rowe, Peter H. Wiernik, Janis Racevskis, Elisabeth Paietta, Alberto Ambesi-Impiobato, Michael Hadler, Mireia Castillo-Martin, and Jeremy B. Samon
- Abstract
PDF file, 69KB.
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Data from Phf6 Loss Enhances HSC Self-Renewal Driving Tumor Initiation and Leukemia Stem Cell Activity in T-ALL
- Author
-
Adolfo A. Ferrando, Pieter Van Vlierberghe, Raul Rabadan, Mignon L. Loh, Julie M. Gastier-Foster, Giuseppe Basso, Maddalena Paganin, Elisabeth Paietta, Martin S. Tallman, Mark R. Litzow, Alberto Ambesi-Impiombato, Chioma J. Madubata, Marissa Rashkovan, S. Aidan Quinn, and Agnieszka A. Wendorff
- Abstract
Supplementary Figures 1-8
- Published
- 2023
- Full Text
- View/download PDF
14. Data from Enhancer Hijacking Drives Oncogenic BCL11B Expression in Lineage-Ambiguous Stem Cell Leukemia
- Author
-
Charles G. Mullighan, Claudia Haferlach, Jeffery M. Klco, John E. Dick, Gang Wu, Torsten Haferlach, Wolfgang Kern, Jinghui Zhang, Adolfo A. Ferrando, Elisabeth M. Paietta, Wendy Stock, William E. Evans, Mary V. Relling, Jun J. Yang, Kathryn G. Roberts, Kristine R. Crews, Wenjian Yang, Ching-Hon Pui, Mignon L. Loh, Stephen P. Hunger, Steven M. Kornblau, Jun Yin, Victoria Wang, Monique L. den Boer, Jacob M. Rowe, Mark R. Litzow, Selina Luger, Marissa Rashkovan, Marcus B. Valentine, Chunxu Qu, Shunsuke Kimura, Anna Stengel, Jing Ma, Tamara Westover, Ti-Cheng Chang, Zhongshan Cheng, Beisi Xu, Cyrus M. Mehr, Paul E. Mead, Ryan Hiltenbrand, Sherif Abdelhamed, Ilaria Iacobucci, Kirsten Dickerson, Laura Garcia-Prat, Andy Zeng, Alex Murison, Xiaotu Ma, Petri Pölönen, Xiaolong Chen, Zhaohui Gu, Sonja Bendig, and Lindsey E. Montefiori
- Abstract
Lineage-ambiguous leukemias are high-risk malignancies of poorly understood genetic basis. Here, we describe a distinct subgroup of acute leukemia with expression of myeloid, T lymphoid, and stem cell markers driven by aberrant allele-specific deregulation of BCL11B, a master transcription factor responsible for thymic T-lineage commitment and specification. Mechanistically, this deregulation was driven by chromosomal rearrangements that juxtapose BCL11B to superenhancers active in hematopoietic progenitors, or focal amplifications that generate a superenhancer from a noncoding element distal to BCL11B. Chromatin conformation analyses demonstrated long-range interactions of rearranged enhancers with the expressed BCL11B allele and association of BCL11B with activated hematopoietic progenitor cell cis-regulatory elements, suggesting BCL11B is aberrantly co-opted into a gene regulatory network that drives transformation by maintaining a progenitor state. These data support a role for ectopic BCL11B expression in primitive hematopoietic cells mediated by enhancer hijacking as an oncogenic driver of human lineage-ambiguous leukemia.Significance:Lineage-ambiguous leukemias pose significant diagnostic and therapeutic challenges due to a poorly understood molecular and cellular basis. We identify oncogenic deregulation of BCL11B driven by diverse structural alterations, including de novo superenhancer generation, as the driving feature of a subset of lineage-ambiguous leukemias that transcend current diagnostic boundaries.This article is highlighted in the In This Issue feature, p. 2659
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Figure 3 from Preclinical Analysis of the γ-Secretase Inhibitor PF-03084014 in Combination with Glucocorticoids in T-cell Acute Lymphoblastic Leukemia
- Author
-
Adolfo A. Ferrando, Carlos Cordon-Cardo, Sophia Randolph, John Jakubczak, Jacob M. Rowe, Peter H. Wiernik, Janis Racevskis, Elisabeth Paietta, Alberto Ambesi-Impiobato, Michael Hadler, Mireia Castillo-Martin, and Jeremy B. Samon
- Abstract
PDF file, 306KB, Interaction between PF-03084014 and chemotherapeutic agents.
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Figure 1 from Preclinical Analysis of the γ-Secretase Inhibitor PF-03084014 in Combination with Glucocorticoids in T-cell Acute Lymphoblastic Leukemia
- Author
-
Adolfo A. Ferrando, Carlos Cordon-Cardo, Sophia Randolph, John Jakubczak, Jacob M. Rowe, Peter H. Wiernik, Janis Racevskis, Elisabeth Paietta, Alberto Ambesi-Impiobato, Michael Hadler, Mireia Castillo-Martin, and Jeremy B. Samon
- Abstract
PDF file, 252KB, PF-03084014 reverses glucocorticoid resistance in T-ALL cell lines.
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Tables from Enhancer Hijacking Drives Oncogenic BCL11B Expression in Lineage-Ambiguous Stem Cell Leukemia
- Author
-
Charles G. Mullighan, Claudia Haferlach, Jeffery M. Klco, John E. Dick, Gang Wu, Torsten Haferlach, Wolfgang Kern, Jinghui Zhang, Adolfo A. Ferrando, Elisabeth M. Paietta, Wendy Stock, William E. Evans, Mary V. Relling, Jun J. Yang, Kathryn G. Roberts, Kristine R. Crews, Wenjian Yang, Ching-Hon Pui, Mignon L. Loh, Stephen P. Hunger, Steven M. Kornblau, Jun Yin, Victoria Wang, Monique L. den Boer, Jacob M. Rowe, Mark R. Litzow, Selina Luger, Marissa Rashkovan, Marcus B. Valentine, Chunxu Qu, Shunsuke Kimura, Anna Stengel, Jing Ma, Tamara Westover, Ti-Cheng Chang, Zhongshan Cheng, Beisi Xu, Cyrus M. Mehr, Paul E. Mead, Ryan Hiltenbrand, Sherif Abdelhamed, Ilaria Iacobucci, Kirsten Dickerson, Laura Garcia-Prat, Andy Zeng, Alex Murison, Xiaotu Ma, Petri Pölönen, Xiaolong Chen, Zhaohui Gu, Sonja Bendig, and Lindsey E. Montefiori
- Abstract
This file contains Supplementary Tables 1-15 detailing clinical and genomic information about each patient analyzed in this study, along with the results of RNA-seq and gene set enrichment analyses.
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Figures, and Supplementary Table Legends from Enhancer Hijacking Drives Oncogenic BCL11B Expression in Lineage-Ambiguous Stem Cell Leukemia
- Author
-
Charles G. Mullighan, Claudia Haferlach, Jeffery M. Klco, John E. Dick, Gang Wu, Torsten Haferlach, Wolfgang Kern, Jinghui Zhang, Adolfo A. Ferrando, Elisabeth M. Paietta, Wendy Stock, William E. Evans, Mary V. Relling, Jun J. Yang, Kathryn G. Roberts, Kristine R. Crews, Wenjian Yang, Ching-Hon Pui, Mignon L. Loh, Stephen P. Hunger, Steven M. Kornblau, Jun Yin, Victoria Wang, Monique L. den Boer, Jacob M. Rowe, Mark R. Litzow, Selina Luger, Marissa Rashkovan, Marcus B. Valentine, Chunxu Qu, Shunsuke Kimura, Anna Stengel, Jing Ma, Tamara Westover, Ti-Cheng Chang, Zhongshan Cheng, Beisi Xu, Cyrus M. Mehr, Paul E. Mead, Ryan Hiltenbrand, Sherif Abdelhamed, Ilaria Iacobucci, Kirsten Dickerson, Laura Garcia-Prat, Andy Zeng, Alex Murison, Xiaotu Ma, Petri Pölönen, Xiaolong Chen, Zhaohui Gu, Sonja Bendig, and Lindsey E. Montefiori
- Abstract
This file contains Supplementary Figures 1-23 with corresponding supplementary figure legends, as well as the legends for Supplementary Tables 1-15.
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Figure 2 from Preclinical Analysis of the γ-Secretase Inhibitor PF-03084014 in Combination with Glucocorticoids in T-cell Acute Lymphoblastic Leukemia
- Author
-
Adolfo A. Ferrando, Carlos Cordon-Cardo, Sophia Randolph, John Jakubczak, Jacob M. Rowe, Peter H. Wiernik, Janis Racevskis, Elisabeth Paietta, Alberto Ambesi-Impiobato, Michael Hadler, Mireia Castillo-Martin, and Jeremy B. Samon
- Abstract
PDF file, 567KB, Absence of synergistic interaction between PF-03084014 and glucocorticoids in gamma-secretase inhibitor- and glucocorticoid-resistant cell lines.
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Methods and Figures from PRC2 Inhibitors Overcome Glucocorticoid Resistance Driven by NSD2 Mutation in Pediatric Acute Lymphoblastic Leukemia
- Author
-
Jonathan D. Licht, Richard B. Lock, Matthew D. Hall, Adolfo A. Ferrando, Richard L. Bennett, Alberto Riva, Min Shen, Christine M. Will, Jon A. Oyer, Marta Kulis, Alok Swaroop, Catalina Troche, Heidi L. Casellas Román, Duohui Jing, Jacob S. Roth, Daphne Dupéré-Richér, Crissandra Piper, Jonathan H. Shrimp, Julia Hlavka-Zhang, and Jianping Li
- Abstract
Supplementary Methods and Figures
- Published
- 2023
- Full Text
- View/download PDF
21. Data from PRC2 Inhibitors Overcome Glucocorticoid Resistance Driven by NSD2 Mutation in Pediatric Acute Lymphoblastic Leukemia
- Author
-
Jonathan D. Licht, Richard B. Lock, Matthew D. Hall, Adolfo A. Ferrando, Richard L. Bennett, Alberto Riva, Min Shen, Christine M. Will, Jon A. Oyer, Marta Kulis, Alok Swaroop, Catalina Troche, Heidi L. Casellas Román, Duohui Jing, Jacob S. Roth, Daphne Dupéré-Richér, Crissandra Piper, Jonathan H. Shrimp, Julia Hlavka-Zhang, and Jianping Li
- Abstract
Mutations in epigenetic regulators are common in relapsed pediatric acute lymphoblastic leukemia (ALL). Here, we uncovered the mechanism underlying the relapse of ALL driven by an activating mutation of the NSD2 histone methyltransferase (p.E1099K). Using high-throughput drug screening, we found that NSD2-mutant cells were specifically resistant to glucocorticoids. Correction of this mutation restored glucocorticoid sensitivity. The transcriptional response to glucocorticoids was blocked in NSD2-mutant cells due to depressed glucocorticoid receptor (GR) levels and the failure of glucocorticoids to autoactivate GR expression. Although H3K27me3 was globally decreased by NSD2 p.E1099K, H3K27me3 accumulated at the NR3C1 (GR) promoter. Pretreatment of NSD2 p.E1099K cell lines and patient-derived xenograft samples with PRC2 inhibitors reversed glucocorticoid resistance in vitro and in vivo. PRC2 inhibitors restored NR3C1 autoactivation by glucocorticoids, increasing GR levels and allowing GR binding and activation of proapoptotic genes. These findings suggest a new therapeutic approach to relapsed ALL associated with NSD2 mutation.Significance:NSD2 histone methyltransferase mutations observed in relapsed pediatric ALL drove glucocorticoid resistance by repression of the GR and abrogation of GR gene autoactivation due to accumulation of K3K27me3 at its promoter. Pretreatment with PRC2 inhibitors reversed resistance, suggesting a new therapeutic approach to these patients with ALL.This article is highlighted in the In This Issue feature, p. 1
- Published
- 2023
- Full Text
- View/download PDF
22. Data from Preclinical Analysis of the γ-Secretase Inhibitor PF-03084014 in Combination with Glucocorticoids in T-cell Acute Lymphoblastic Leukemia
- Author
-
Adolfo A. Ferrando, Carlos Cordon-Cardo, Sophia Randolph, John Jakubczak, Jacob M. Rowe, Peter H. Wiernik, Janis Racevskis, Elisabeth Paietta, Alberto Ambesi-Impiobato, Michael Hadler, Mireia Castillo-Martin, and Jeremy B. Samon
- Abstract
T-cell acute lymphoblastic leukemias (T-ALL) and lymphomas are aggressive hematologic cancers frequently associated with activating mutations in NOTCH1. Early studies identified NOTCH1 as an attractive therapeutic target for the treatment of T-ALL through the use of γ-secretase inhibitors (GSI). Here, we characterized the interaction between PF-03084014, a clinically relevant GSI, and dexamethasone in preclinical models of glucocorticoid-resistant T-ALL. Combination treatment of the GSI PF-03084014 with glucocorticoids induced a synergistic antileukemic effect in human T-ALL cell lines and primary human T-ALL patient samples. Mechanistically PF-03084014 plus glucocorticoid treatment induced increased transcriptional upregulation of the glucocorticoid receptor and glucocorticoid target genes. Treatment with PF-03084014 and glucocorticoids in combination was highly efficacious in vivo, with enhanced reduction of tumor burden in a xenograft model of T-ALL. Finally, glucocorticoid treatment effectively reversed PF-03084014–induced gastrointestinal toxicity via inhibition of goblet cell metaplasia. These results warrant the analysis of PF-03084014 and glucocorticoids in combination for the treatment of glucocorticoid-resistant T-ALL. Mol Cancer Ther; 11(7); 1565–75. ©2012 AACR.
- Published
- 2023
- Full Text
- View/download PDF
23. Data from Phf6 Loss Enhances HSC Self-Renewal Driving Tumor Initiation and Leukemia Stem Cell Activity in T-ALL
- Author
-
Adolfo A. Ferrando, Pieter Van Vlierberghe, Raul Rabadan, Mignon L. Loh, Julie M. Gastier-Foster, Giuseppe Basso, Maddalena Paganin, Elisabeth Paietta, Martin S. Tallman, Mark R. Litzow, Alberto Ambesi-Impiombato, Chioma J. Madubata, Marissa Rashkovan, S. Aidan Quinn, and Agnieszka A. Wendorff
- Abstract
The plant homeodomain 6 gene (PHF6) is frequently mutated in human T-cell acute lymphoblastic leukemia (T-ALL); however, its specific functional role in leukemia development remains to be established. Here, we show that loss of PHF6 is an early mutational event in leukemia transformation. Mechanistically, genetic inactivation of Phf6 in the hematopoietic system enhances hematopoietic stem cell (HSC) long-term self-renewal and hematopoietic recovery after chemotherapy by rendering Phf6 knockout HSCs more quiescent and less prone to stress-induced activation. Consistent with a leukemia-initiating tumor suppressor role, inactivation of Phf6 in hematopoietic progenitors lowers the threshold for the development of NOTCH1-induced T-ALL. Moreover, loss of Phf6 in leukemia lymphoblasts activates a leukemia stem cell transcriptional program and drives enhanced T-ALL leukemia-initiating cell activity. These results implicate Phf6 in the control of HSC homeostasis and long-term self-renewal and support a role for PHF6 loss as a driver of leukemia-initiating cell activity in T-ALL.Significance:Phf6 controls HSC homeostasis, leukemia initiation, and T-ALL leukemia-initiating cell self-renewal. These results substantiate a role for PHF6 mutations as early events and drivers of leukemia stem cell activity in the pathogenesis of T-ALL.This article is highlighted in the In This Issue feature, p. 305
- Published
- 2023
- Full Text
- View/download PDF
24. Targeting S100A9–ALDH1A1–Retinoic Acid Signaling to Suppress Brain Relapse in EGFR-Mutant Lung Cancer
- Author
-
Anup Kumar Biswas, Seoyoung Han, Yifan Tai, Wanchao Ma, Courtney Coker, S. Aidan Quinn, Ahmad Rushdi Shakri, Timothy James Zhong, Hanna Scholze, Galina G. Lagos, Angeliki Mela, Katia Manova-Todorova, Elisa de Stanchina, Adolfo A. Ferrando, Cathy Mendelsohn, Peter Canoll, Helena A. Yu, Paul K. Paik, Anjali Saqi, Catherine A. Shu, Mark G. Kris, Joan Massague, and Swarnali Acharyya
- Subjects
Aniline Compounds ,Lung Neoplasms ,Brain ,Retinal Dehydrogenase ,Tretinoin ,Article ,Aldehyde Dehydrogenase 1 Family ,ErbB Receptors ,Oncology ,Carcinoma, Non-Small-Cell Lung ,Mutation ,Humans ,Neoplasm Recurrence, Local ,Protein Kinase Inhibitors ,Signal Transduction - Abstract
The epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) osimertinib has significantly prolonged progression-free survival (PFS) in patients with EGFR-mutant lung cancer, including those with brain metastases. However, despite striking initial responses, osimertinib-treated patients eventually develop lethal metastatic relapse, often to the brain. Although osimertinib-refractory brain relapse is a major clinical challenge, its underlying mechanisms remain poorly understood. Using metastatic models of EGFR-mutant lung cancer, we show that cancer cells expressing high intracellular S100A9 escape osimertinib and initiate brain relapses. Mechanistically, S100A9 upregulates ALDH1A1 expression and activates the retinoic acid (RA) signaling pathway in osimertinib-refractory cancer cells. We demonstrate that the genetic repression of S100A9, ALDH1A1, or RA receptors (RAR) in cancer cells, or treatment with a pan-RAR antagonist, dramatically reduces brain metastasis. Importantly, S100A9 expression in cancer cells correlates with poor PFS in osimertinib-treated patients. Our study, therefore, identifies a novel, therapeutically targetable S100A9–ALDH1A1–RA axis that drives brain relapse. Significance: Treatment with the EGFR TKI osimertinib prolongs the survival of patients with EGFR-mutant lung cancer; however, patients develop metastatic relapses, often to the brain. We identified a novel intracellular S100A9–ALDH1A1–RA signaling pathway that drives lethal brain relapse and can be targeted by pan-RAR antagonists to prevent cancer progression and prolong patient survival. This article is highlighted in the In This Issue feature, p. 873
- Published
- 2022
- Full Text
- View/download PDF
25. PRC2 Inhibitors Overcome Glucocorticoid Resistance Driven by NSD2 Mutation in Pediatric Acute Lymphoblastic Leukemia
- Author
-
Crissandra Piper, Matthew D. Hall, Marta Kulis, Richard L. Bennett, Alok Swaroop, Min Shen, Richard B. Lock, Jonathan H. Shrimp, Jon A. Oyer, Christine Will, Alberto Riva, Heidi L. Casellas Roman, Duohui Jing, Jianping Li, Catalina Troche, Adolfo A. Ferrando, Jacob S. Roth, Daphné Dupéré-Richer, Jonathan D. Licht, and Julia Cathryn Hlavka-Zhang
- Subjects
Mutation ,biology ,business.industry ,medicine.disease_cause ,Glucocorticoid receptor ,Glucocorticoid Sensitivity ,Oncology ,In vivo ,Cell culture ,Histone methyltransferase ,medicine ,Cancer research ,biology.protein ,Epigenetics ,PRC2 ,business ,hormones, hormone substitutes, and hormone antagonists - Abstract
Mutations in epigenetic regulators are common in relapsed pediatric acute lymphoblastic leukemia (ALL). Here, we uncovered the mechanism underlying the relapse of ALL driven by an activating mutation of the NSD2 histone methyltransferase (p.E1099K). Using high-throughput drug screening, we found that NSD2-mutant cells were specifically resistant to glucocorticoids. Correction of this mutation restored glucocorticoid sensitivity. The transcriptional response to glucocorticoids was blocked in NSD2-mutant cells due to depressed glucocorticoid receptor (GR) levels and the failure of glucocorticoids to autoactivate GR expression. Although H3K27me3 was globally decreased by NSD2 p.E1099K, H3K27me3 accumulated at the NR3C1 (GR) promoter. Pretreatment of NSD2 p.E1099K cell lines and patient-derived xenograft samples with PRC2 inhibitors reversed glucocorticoid resistance in vitro and in vivo. PRC2 inhibitors restored NR3C1 autoactivation by glucocorticoids, increasing GR levels and allowing GR binding and activation of proapoptotic genes. These findings suggest a new therapeutic approach to relapsed ALL associated with NSD2 mutation. Significance: NSD2 histone methyltransferase mutations observed in relapsed pediatric ALL drove glucocorticoid resistance by repression of the GR and abrogation of GR gene autoactivation due to accumulation of K3K27me3 at its promoter. Pretreatment with PRC2 inhibitors reversed resistance, suggesting a new therapeutic approach to these patients with ALL. This article is highlighted in the In This Issue feature, p. 1
- Published
- 2022
- Full Text
- View/download PDF
26. Supplementary figure legends from Tumor Suppressor HIPK2 Regulates Malignant Growth via Phosphorylation of Notch1
- Author
-
Hee-Sae Park, Keesook Lee, Adolfo A. Ferrando, Cheol Yong Choi, Ji Shin Lee, Kyung-Hee Chun, Dong Wook Choi, Hyeok-Gu Kang, Hyun-Woo Lee, Hye-Jin Lee, Eun-Hye Jo, Ji-Seon Ahn, Ji-Hye Yoon, Mi-Yeon Kim, and Eun-Jung Ann
- Abstract
Supplementary figure legends
- Published
- 2023
- Full Text
- View/download PDF
27. Supplementary table 1 and 2 from Tumor Suppressor HIPK2 Regulates Malignant Growth via Phosphorylation of Notch1
- Author
-
Hee-Sae Park, Keesook Lee, Adolfo A. Ferrando, Cheol Yong Choi, Ji Shin Lee, Kyung-Hee Chun, Dong Wook Choi, Hyeok-Gu Kang, Hyun-Woo Lee, Hye-Jin Lee, Eun-Hye Jo, Ji-Seon Ahn, Ji-Hye Yoon, Mi-Yeon Kim, and Eun-Jung Ann
- Abstract
Supplementary Table S1: Immunohistochemical staining of Notch1-IC and HIPK2 in serial tissue arrays of 61 breast cancer samples. Supplementary Table S2: Semi-Quantitative analysis of Notch1-IC, phosphorylated Notch1-IC T2512, HIPK2, and Fbw7 expression in breast cancer tissues.
- Published
- 2023
- Full Text
- View/download PDF
28. Supplementary Materials and Methods from Tumor Suppressor HIPK2 Regulates Malignant Growth via Phosphorylation of Notch1
- Author
-
Hee-Sae Park, Keesook Lee, Adolfo A. Ferrando, Cheol Yong Choi, Ji Shin Lee, Kyung-Hee Chun, Dong Wook Choi, Hyeok-Gu Kang, Hyun-Woo Lee, Hye-Jin Lee, Eun-Hye Jo, Ji-Seon Ahn, Ji-Hye Yoon, Mi-Yeon Kim, and Eun-Jung Ann
- Abstract
Description of additional methods and procedures used in the study.
- Published
- 2023
- Full Text
- View/download PDF
29. Supplementary figures S1-10 from Tumor Suppressor HIPK2 Regulates Malignant Growth via Phosphorylation of Notch1
- Author
-
Hee-Sae Park, Keesook Lee, Adolfo A. Ferrando, Cheol Yong Choi, Ji Shin Lee, Kyung-Hee Chun, Dong Wook Choi, Hyeok-Gu Kang, Hyun-Woo Lee, Hye-Jin Lee, Eun-Hye Jo, Ji-Seon Ahn, Ji-Hye Yoon, Mi-Yeon Kim, and Eun-Jung Ann
- Abstract
Supplementary Figure 1: ADR suppresses Notch1 signaling. Supplementary Figure 2: ADR inhibits the transcriptional activity of Notch1 by inducing HIPK2. Supplementary Figure 3: HIPK2 regulates the Notch1-IC protein stability through proteasomal degradation pathways. Supplementary Figure 4: HIPK2 facilitates Notch1-IC degradation by Fbw7 ubiquitin ligase. Supplementary Figure 5: HIPK2 physically interacts with Notch1-IC. Supplementary Figure 6: HIPK2 phosphorylates Notch1-IC at the T2512 residue. Supplementary Figure 7: HIPK2-mediated downregulation of Notch1-IC suppresses the tumorigenesis of breast cancer. Supplementary Figure 8: Somatic mutations in the CPD motif of Notch1-IC make it resistant to HIPK2. Supplementary Figure 9: Physiological effects of somatic mutations in the CPD motif of Notch1-IC. Supplementary Figure 10: A propose model of Notch1 signaling pathway regulation by HIPK2 in cancer during chemotherapy.
- Published
- 2023
- Full Text
- View/download PDF
30. Data from Tumor Suppressor HIPK2 Regulates Malignant Growth via Phosphorylation of Notch1
- Author
-
Hee-Sae Park, Keesook Lee, Adolfo A. Ferrando, Cheol Yong Choi, Ji Shin Lee, Kyung-Hee Chun, Dong Wook Choi, Hyeok-Gu Kang, Hyun-Woo Lee, Hye-Jin Lee, Eun-Hye Jo, Ji-Seon Ahn, Ji-Hye Yoon, Mi-Yeon Kim, and Eun-Jung Ann
- Abstract
The receptor Notch1 plays an important role in malignant progression of many cancers, but its regulation is not fully understood. In this study, we report that the kinase HIPK2 is responsible for facilitating the Fbw7-dependent proteasomal degradation of Notch1 by phosphorylating its intracellular domain (Notch1-IC) within the Cdc4 phosphodegron motif. Notch1-IC expression was higher in cancer cells than normal cells. Under genotoxic stress, Notch1-IC was phosphorylated constitutively by HIPK2 and was maintained at a low level through proteasomal degradation. HIPK2 phosphorylated the residue T2512 in Notch1-IC. Somatic mutations near this residue rendered Notch1-IC resistant to degradation, as induced either by HIPK2 overexpression or adriamycin treatment. In revealing an important mechanism of Notch1 stability, the results of this study could offer a therapeutic strategy to block Notch1-dependent progression in many types of cancer. Cancer Res; 76(16); 4728–40. ©2016 AACR.
- Published
- 2023
- Full Text
- View/download PDF
31. Pharmacological inhibition of NT5C2 reverses genetic and non-genetic drivers of 6-MP resistance in acute lymphoblastic leukemia
- Author
-
Clara Reglero, Chelsea L. Dieck, Arie Zask, Farhad Forouhar, Anouchka P. Laurent, Wen-Hsuan W. Lin, Robert Albero, Hannah I. Miller, Cindy Ma, Julie M. Gastier-Foster, Mignon L. Loh, Liang Tong, Brent R. Stockwell, Teresa Palomero, and Adolfo A. Ferrando
- Subjects
Oncology ,Mercaptopurine ,Drug Resistance, Neoplasm ,Recurrence ,Humans ,Antineoplastic Agents ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,5'-Nucleotidase ,Article - Abstract
Low-intensity maintenance therapy with 6-mercaptopurine (6-MP) limits the occurrence of acute lymphoblastic leukemia (ALL) relapse and is central to the success of multiagent chemotherapy protocols. Activating mutations in the 5′-nucleotidase cytosolic II (NT5C2) gene drive resistance to 6-MP in over 35% of early relapse ALL cases. Here we identify CRCD2 as a first-in-class small-molecule NT5C2 nucleotidase inhibitor broadly active against leukemias bearing highly prevalent relapse-associated mutant forms of NT5C2 in vitro and in vivo. Importantly, CRCD2 treatment also enhanced the cytotoxic activity of 6-MP in NT5C2 wild-type leukemias, leading to the identification of NT5C2 Ser502 phosphorylation as a novel NT5C2-mediated mechanism of 6-MP resistance in this disease. These results uncover an unanticipated role of nongenetic NT5C2 activation as a driver of 6-MP resistance in ALL and demonstrate the potential of NT5C2 inhibitor therapy for enhancing the efficacy of thiopurine maintenance therapy and overcoming resistance at relapse. Significance: Relapse-associated NT5C2 mutations directly contribute to relapse in ALL by driving resistance to chemotherapy with 6-MP. Pharmacologic inhibition of NT5C2 with CRCD2, a first-in-class nucleotidase inhibitor, enhances the cytotoxic effects of 6-MP and effectively reverses thiopurine resistance mediated by genetic and nongenetic mechanisms of NT5C2 activation in ALL. This article is highlighted in the In This Issue feature, p. 2483
- Published
- 2022
32. Insights into the mechanisms underlying aberrant SOX11 oncogene expression in mantle cell lymphoma
- Author
-
Marc A. Marti-Renom, María José Calasanz, Núria Verdaguer-Dot, Xabier Agirre, Dolors Colomer, Maribel Parra, Roser Vilarrasa-Blasi, Paula Soler-Vila, Ana C. Queirós, Vicente Chapaprieta, Adolfo A. Ferrando, Elias Campo, Marta Kulis, José I. Martín-Subero, Felipe Prosper, Laura Belver, Renée Beekman, and Sílvia Beà
- Subjects
Cancer Research ,Oncogene ,business.industry ,Lymphoma, Mantle-Cell ,Hematology ,Biology ,Chromatin Assembly and Disassembly ,medicine.disease ,SOXC Transcription Factors ,Enhancer Elements, Genetic ,Text mining ,Oncology ,Expression (architecture) ,Tumor Cells, Cultured ,medicine ,Cancer research ,Humans ,Mantle cell lymphoma ,Promoter Regions, Genetic ,B-cell lymphoma ,business - Published
- 2021
- Full Text
- View/download PDF
33. Enhancer Hijacking Drives Oncogenic BCL11B Expression in Lineage-Ambiguous Stem Cell Leukemia
- Author
-
Jeffery M. Klco, Jing Ma, Xiaotu Ma, Ryan Hiltenbrand, Mary V. Relling, William E. Evans, Petri Pölönen, Paul E. Mead, Sonja Bendig, Steven M. Kornblau, Gang Wu, Selina M. Luger, Mark R. Litzow, Jacob M. Rowe, Stephen P. Hunger, Tamara Westover, Ti-Cheng Chang, Shunsuke Kimura, Jinghui Zhang, Elisabeth Paietta, Jun J. Yang, Alex Murison, Laura García-Prat, Torsten Haferlach, Ilaria Iacobucci, Chunxu Qu, Zhaohui Gu, Mignon L. Loh, Lindsey E. Montefiori, Marissa Rashkovan, Wenjian Yang, Marcus B. Valentine, Kathryn G. Roberts, Zhongshan Cheng, Beisi Xu, Victoria Wang, Jun Yin, Wendy Stock, Kirsten Dickerson, Cyrus M. Mehr, Claudia Haferlach, Adolfo A. Ferrando, Wolfgang Kern, Ching-Hon Pui, Kristine R. Crews, Andy G.X. Zeng, Sherif Abdelhamed, John E. Dick, Xiao-Long Chen, Anna Stengel, Charles G. Mullighan, and Monique L. den Boer
- Subjects
Myeloid ,BCL11B ,Biology ,medicine.disease ,Stem cell marker ,Haematopoiesis ,Leukemia ,medicine.anatomical_structure ,Oncology ,medicine ,Cancer research ,Progenitor cell ,Enhancer ,Transcription factor - Abstract
Lineage-ambiguous leukemias are high-risk malignancies of poorly understood genetic basis. Here, we describe a distinct subgroup of acute leukemia with expression of myeloid, T lymphoid, and stem cell markers driven by aberrant allele-specific deregulation of BCL11B, a master transcription factor responsible for thymic T-lineage commitment and specification. Mechanistically, this deregulation was driven by chromosomal rearrangements that juxtapose BCL11B to superenhancers active in hematopoietic progenitors, or focal amplifications that generate a superenhancer from a noncoding element distal to BCL11B. Chromatin conformation analyses demonstrated long-range interactions of rearranged enhancers with the expressed BCL11B allele and association of BCL11B with activated hematopoietic progenitor cell cis-regulatory elements, suggesting BCL11B is aberrantly co-opted into a gene regulatory network that drives transformation by maintaining a progenitor state. These data support a role for ectopic BCL11B expression in primitive hematopoietic cells mediated by enhancer hijacking as an oncogenic driver of human lineage-ambiguous leukemia. Significance: Lineage-ambiguous leukemias pose significant diagnostic and therapeutic challenges due to a poorly understood molecular and cellular basis. We identify oncogenic deregulation of BCL11B driven by diverse structural alterations, including de novo superenhancer generation, as the driving feature of a subset of lineage-ambiguous leukemias that transcend current diagnostic boundaries. This article is highlighted in the In This Issue feature, p. 2659
- Published
- 2021
- Full Text
- View/download PDF
34. Covalent inhibition of NSD1 histone methyltransferase
- Author
-
Sergei Zari, Juliano Ndoj, Jolanta Grembecka, Paula González-Alonso, Katarzyna Kempińska, Mykhaylo A. Potopnyk, Tomasz Cierpicki, Hongzhi Miao, Caroline Nikolaidis, Andrew D. Henderson, Huang Huang, Trupta Purohit, David S. Rogawski, Shirish Shukla, Christina A. Howard, Adolfo A. Ferrando, Hyo Je Cho, Joshua Abbott, Anna K. Mapp, Maria Luisa Sulis, and Hao Li
- Subjects
Models, Molecular ,Protein Conformation, alpha-Helical ,Conformational change ,Oncogene Proteins, Fusion ,Antineoplastic Agents ,Plasma protein binding ,Substrate Specificity ,03 medical and health sciences ,Histone H3 ,Protein structure ,Leukocytes ,Tumor Cells, Cultured ,Humans ,Protein Interaction Domains and Motifs ,Enzyme Inhibitors ,Myeloid Ecotropic Viral Integration Site 1 Protein ,Molecular Biology ,030304 developmental biology ,Homeodomain Proteins ,0303 health sciences ,Binding Sites ,Leukemia ,Gene Expression Regulation, Leukemic ,Chemistry ,030302 biochemistry & molecular biology ,Histone-Lysine N-Methyltransferase ,Cell Biology ,Ligand (biochemistry) ,Small molecule ,Cell biology ,Nuclear Pore Complex Proteins ,Kinetics ,Nuclear receptor ,Histone methyltransferase ,Protein Conformation, beta-Strand ,Protein Binding ,Signal Transduction - Abstract
The nuclear receptor-binding SET domain (NSD) family of histone methyltransferases is associated with various malignancies, including aggressive acute leukemia with NUP98-NSD1 translocation. While NSD proteins represent attractive drug targets, their catalytic SET domains exist in autoinhibited conformation, presenting notable challenges for inhibitor development. Here, we employed a fragment-based screening strategy followed by chemical optimization, which resulted in the development of the first-in-class irreversible small-molecule inhibitors of the nuclear receptor-binding SET domain protein 1 (NSD1) SET domain. The crystal structure of NSD1 in complex with covalently bound ligand reveals a conformational change in the autoinhibitory loop of the SET domain and formation of a channel-like pocket suitable for targeting with small molecules. Our covalent lead-compound BT5-demonstrates on-target activity in NUP98-NSD1 leukemia cells, including inhibition of histone H3 lysine 36 dimethylation and downregulation of target genes, and impaired colony formation in an NUP98-NSD1 patient sample. This study will facilitate the development of the next generation of potent and selective inhibitors of the NSD histone methyltransferases.
- Published
- 2020
- Full Text
- View/download PDF
35. Combinatorial ETS1-Dependent Control of Oncogenic NOTCH1 Enhancers in T-cell Leukemia
- Author
-
Yiran Liu, Cher Sha, Alberto Ambesi-Impiombato, Michael C. Ostrowski, Erin Kim, Theresa M. Keeley, Jahnavi K. Nalamolu, Qing Wang, Mark Y. Chiang, Adolfo A. Ferrando, Ran Yan, Rohan Kodgule, Russell J.H. Ryan, Arvind Rao, Nicholas Kunnath, Linda C. Samuelson, Barbara L. Kee, Giusy Della Gatta, Anna C. McCarter, Rork Kuick, Ashley Melnick, and Mengxi Sun
- Subjects
Leukemia, T-Cell ,Carcinogenesis ,Effector ,Lymphoblastic Leukemia ,T-cell leukemia ,General Medicine ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Article ,Chromatin ,Proto-Oncogene Protein c-ets-1 ,Mice ,ETS1 ,Antineoplastic Combined Chemotherapy Protocols ,Cancer research ,Animals ,Receptor, Notch1 ,Enhancer ,Gene ,Transcription factor ,Signal Transduction - Abstract
Notch activation is highly prevalent among cancers, in particular T-cell acute lymphoblastic leukemia (T-ALL). However, the use of pan-Notch inhibitors to treat cancers has been hampered by adverse effects, particularly intestinal toxicities. To circumvent this barrier in T-ALL, we aimed to inhibit ETS1, a developmentally important T-cell transcription factor previously shown to cobind Notch response elements. Using complementary genetic approaches in mouse models, we show that ablation of Ets1 leads to strong Notch-mediated suppressive effects on T-cell development and leukemogenesis but milder intestinal effects than pan-Notch inhibitors. Mechanistically, genome-wide chromatin profiling studies demonstrate that Ets1 inactivation impairs recruitment of multiple Notch-associated factors and Notch-dependent activation of transcriptional elements controlling major Notch-driven oncogenic effector pathways. These results uncover previously unrecognized hierarchical heterogeneity of Notch-controlled genes and point to Ets1-mediated enucleation of Notch–Rbpj transcriptional complexes as a target for developing specific anti-Notch therapies in T-ALL that circumvent the barriers of pan-Notch inhibition. Significance: Notch signaling controls developmentally important and tissue-specific activities, raising barriers for developing anti-Notch therapies. Pivoting away from pan-Notch inhibitors, we show antileukemic but less toxic effects of targeting ETS1, a T-cell NOTCH1 cofactor. These results demonstrate the feasibility of context-dependent suppression of NOTCH1 programs for the treatment of T-ALL. This article is highlighted in the In This Issue feature, p. 127
- Published
- 2020
- Full Text
- View/download PDF
36. Subclonal NT5C2 mutations are associated with poor outcomes after relapse of pediatric acute lymphoblastic leukemia
- Author
-
Arend von Stackelberg, Hossein Khiabanian, Jui Wan Loh, Stefanie Groeneveld-Krentz, Adolfo A. Ferrando, Malwine J. Barz, Annabell Szymansky, Jana Hof, Cornelia Eckert, Kathy Astrahantseff, and Renate Kirschner-Schwabe
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Adolescent ,DNA Mutational Analysis ,Immunology ,Purine analogue ,medicine.disease_cause ,Biochemistry ,law.invention ,Young Adult ,Gene Frequency ,Recurrence ,law ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Young adult ,Allele ,Child ,5'-Nucleotidase ,Allele frequency ,Alleles ,Polymerase chain reaction ,Mutation ,business.industry ,Hazard ratio ,Wild type ,Infant ,Cell Biology ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Prognosis ,Child, Preschool ,Female ,business ,Biomarkers - Abstract
Activating mutations in cytosolic 5′-nucleotidase II (NT5C2) are considered to drive relapse formation in acute lymphoblastic leukemia (ALL) by conferring purine analog resistance. To examine the clinical effects of NT5C2 mutations in relapsed ALL, we analyzed NT5C2 in 455 relapsed B-cell precursor ALL patients treated within the ALL-REZ BFM 2002 relapse trial using sequencing and sensitive allele-specific real-time polymerase chain reaction. We detected 110 NT5C2 mutations in 75 (16.5%) of 455 B-cell precursor ALL relapses. Two-thirds of relapses harbored subclonal mutations and only one-third harbored clonal mutations. Event-free survival after relapse was inferior in patients with relapses with clonal and subclonal NT5C2 mutations compared with those without (19% and 25% vs 53%, P < .001). However, subclonal, but not clonal, NT5C2 mutations were associated with reduced event-free survival in multivariable analysis (hazard ratio, 1.89; 95% confidence interval, 1.28-2.69; P = .001) and with an increased rate of nonresponse to relapse treatment (subclonal 32%, clonal 12%, wild type 9%, P < .001). Nevertheless, 27 (82%) of 33 subclonal NT5C2 mutations became undetectable at the time of nonresponse or second relapse, and in 10 (71%) of 14 patients subclonal NT5C2 mutations were undetectable already after relapse induction treatment. These results show that subclonal NT5C2 mutations define relapses associated with high risk of treatment failure in patients and at the same time emphasize that their role in outcome is complex and goes beyond mutant NT5C2 acting as a targetable driver during relapse progression. Sensitive, prospective identification of NT5C2 mutations is warranted to improve the understanding and treatment of this aggressive ALL relapse subtype.
- Published
- 2020
- Full Text
- View/download PDF
37. Functional mapping of PHF6 complexes in chromatin remodeling, replication dynamics, and DNA repair
- Author
-
Silvia Alvarez, Ana C. da Silva Almeida, Robert Albero, Mayukh Biswas, Angelica Barreto-Galvez, Thomas S. Gunning, Anam Shaikh, Tomas Aparicio, Agnieszka Wendorff, Erich Piovan, Pieter Van Vlierberghe, Steven Gygi, Jean Gautier, Advaitha Madireddy, and Adolfo A. Ferrando
- Subjects
Leukemia ,DNA Repair ,MUTATIONS ,PROTEINS ,Immunology ,Biology and Life Sciences ,RECOMBINATION ,Cell Biology ,Hematology ,Chromatin Assembly and Disassembly ,Biochemistry ,Chromatin ,Nucleosomes ,FRAGILE SITES ,Repressor Proteins ,INSIGHTS ,HEMATOPOIETIC STEM ,FANCONI-ANEMIA ,Medicine and Health Sciences ,FAILURE ,Humans ,DAMAGE RESPONSE ,KAP-1 PHOSPHORYLATION - Abstract
The Plant Homeodomain 6 gene (PHF6) encodes a nucleolar and chromatin-associated leukemia tumor suppressor with proposed roles in transcription regulation. However, specific molecular mechanisms controlled by PHF6 remain rudimentarily understood. Here we show that PHF6 engages multiple nucleosome remodeling protein complexes, including nucleosome remodeling and deacetylase, SWI/SNF and ISWI factors, the replication machinery and DNA repair proteins. Moreover, after DNA damage, PHF6 localizes to sites of DNA injury, and its loss impairs the resolution of DNA breaks, with consequent accumulation of single- and double-strand DNA lesions. Native chromatin immunoprecipitation sequencing analyses show that PHF6 specifically associates with difficult-to-replicate heterochromatin at satellite DNA regions enriched in histone H3 lysine 9 trimethyl marks, and single-molecule locus-specific analyses identify PHF6 as an important regulator of genomic stability at fragile sites. These results extend our understanding of the molecular mechanisms controlling hematopoietic stem cell homeostasis and leukemia transformation by placing PHF6 at the crossroads of chromatin remodeling, replicative fork dynamics, and DNA repair.
- Published
- 2021
38. Phosphoproteomic profiling of T cell acute lymphoblastic leukemia reveals targetable kinases and combination treatment strategies
- Author
-
Valentina, Cordo', Mariska T, Meijer, Rico, Hagelaar, Richard R, de Goeij-de Haas, Vera M, Poort, Alex A, Henneman, Sander R, Piersma, Thang V, Pham, Koichi, Oshima, Adolfo A, Ferrando, Guido J R, Zaman, Connie R, Jimenez, and Jules P P, Meijerink
- Subjects
Cell Line, Tumor ,T-Lymphocytes ,Dasatinib ,Humans ,Phosphorylation ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Protein Kinase Inhibitors - Abstract
Protein kinase inhibitors are amongst the most successful cancer treatments, but targetable kinases activated by genomic abnormalities are rare in T cell acute lymphoblastic leukemia. Nevertheless, kinases can be activated in the absence of genetic defects. Thus, phosphoproteomics can provide information on pathway activation and signaling networks that offer opportunities for targeted therapy. Here, we describe a mass spectrometry-based global phosphoproteomic profiling of 11 T cell acute lymphoblastic leukemia cell lines to identify targetable kinases. We report a comprehensive dataset consisting of 21,000 phosphosites on 4,896 phosphoproteins, including 217 kinases. We identify active Src-family kinases signaling as well as active cyclin-dependent kinases. We validate putative targets for therapy ex vivo and identify potential combination treatments, such as the inhibition of the INSR/IGF-1R axis to increase the sensitivity to dasatinib treatment. Ex vivo validation of selected drug combinations using patient-derived xenografts provides a proof-of-concept for phosphoproteomics-guided design of personalized treatments.
- Published
- 2021
39. Overcoming NOTCH1-Driven Chemoresistance in T-Cell Acute Lymphoblastic Leukemia Via Metabolic Intervention with Oxphos Inhibitor
- Author
-
Katarzyna Tomczak, Katayoun Rezvani, Ken Furudate, Mecit Kaplan, Terzah M. Horton, Helen Ma, Shanti Rojas-Sutterlin, Jiyang Yu, Elias Jabbour, Steven M. Kornblau, Diogo Troggian Veiga, Daniel Herranz, Koichi Takahashi, Jared Henderson, Adolfo A. Ferrando, Yogesh Dhungana, Eric Davis, Trang Hoang, Fieke W Hoff, Alessia Lodi, Anna Skwarska, Shelley M. Herbrich, Maria Emilia Di Francesco, Di Du, Natalia Baran, Stefano Tiziani, Joseph R. Marszalek, Pandey Renu, David T. Teachey, Vivian Ruvolo, Sriram S. Shanmugavelandy, Sujan Piya, Ondrej Havranek, Shannon R. Sweeney, Vinitha Mary Kuruvilla, Philip L. Lorenzi, Ningping Feng, Karine Harutyunyan, Marina Konopleva, Marcin Kamiński, André Haman, Marc O. Warmoes, Mihai Gagea, Michael Andreeff, Jun J. Yang, May Daher, Luciana Melo Garcia, Wentao Yang, and Antonio Cavazos
- Subjects
medicine.anatomical_structure ,business.industry ,Intervention (counseling) ,Lymphoblastic Leukemia ,T cell ,Immunology ,Cancer research ,Medicine ,Cell Biology ,Hematology ,Oxidative phosphorylation ,business ,Biochemistry - Abstract
The inferior cure rate of T-cell acute lymphoblastic leukemia (T-ALL) is associated with inherent drug resistance. The activating NOTCH1 gene mutations have been reported to cause chemoresistance at the stem cell level1. Direct NOTCH1 inhibition has failed in clinical trials due to a narrow therapeutic window but targeting key oncogenic and metabolic pathways downstream of mutated NOTCH1 may offer novel approaches. We previously reported that rapid transformation of thymocytes at the DN3 differentiation stage into preleukemic stem cells (pre-LSC) requires elevated Notch1 in addition to the presence of Scl/Lmo11. Notably, we showed that cellular metabolism of NOTCH1-mutated T-ALLs depends on Oxidative Phosphorylation (OxPhos) and that OxPhos inhibition using the complex I inhibitor IACS-010759 (OxPhos-i) is efficacious in NOTCH1-mutated T-ALL patient derived xenografts (PDXs)2. Here, we investigated the link between NOTCH1-mutated chemoresistance and OxPhos in pre-leukemic and leukemic cells, utilizing comprehensive molecular and functional assays. We hypothesized that chemotherapy aided by OxPhos-i overcomes chemoresistance, depletes LSCs and combats T-ALL. First, we analyzed the role of OxPhos in downstream Notch1 targets at the pre- and leukemic stage considering four stages of thymocyte differentiation (D1-D4), in a mouse model of human T-ALL1. Gene set enrichment analysis (GSEA) implicated increased expression of Notch1 target genes starting at DN1, and OxPhos target genes were the highest-ranked gene set at DN3. Next, activation of Notch1 by its ligand DL4 and inhibition of OxPhos reduced viability of pre-LSCs, indicating that ligand-dependent activation of Notch1 signaling upregulates the OxPhos pathway and sensitizes pre-LSCs to OxPhos-i. To clarify the role of Notch1 signaling, we examined the effect of IACS-010759 on pre-leukemic thymocytes harboring LMO1, SCL-LMO1, NOTCH1, LMO1-NOTCH1 and SCL-LMO1-NOTCH1 with and without DL4 stimulation. We found that in the absence of DL4, only thymocytes harboring the Notch1 oncogene responded to OxPhos-i, whereas all DL4-stimulated thymocytes responded regardless of Notch1 status (Fig. 1a). In addition, at the leukemic stage, we found elevation of the OxPhos pathway driven by oncogenic Notch1 when we compared transcriptomes of SCL-LMO1 induced T-ALL in the presence or absence of the NOTCH1 oncogene. In line with the murine T-ALL NOTCH1 model, we performed transcriptome analysis of two independent T-ALL patient cohorts prior to chemotherapy, COG TARGET ALL (n=263) and AALL1231 (n=75), comparing transcriptomes of NOTCH1-mutated vs NOTCH1-wt T-ALLs. We found co-segregation of NOTCH1 mutations with significant upregulation of OxPhos and TCA cycle genes and downregulation of apoptosis signaling. Aiming to reverse the NOTCH1-controlled anti-apoptotic program and chemoresistance, we next tested the combination of Vincristine, Dexamethasone and L-Asparaginase (VXL) with IACS-010759. When compared to vehicle, OxPhos-i or VXL alone, only the VXL-OxPhos-i treatment caused an energetic crisis indicated by decreased OCR and ECAR (Seahorse), which translated to a profound reduction of viability (CTG, flow cytometry) in T-ALL cell lines (n=9) and primary T-ALL samples (n=5). Additionally, the IACS-VXL combination in vivo resulted in pan-metabolic blockade, which caused metabolic shut-down and triggered early induction of apoptosis in leukemic cells in peripheral blood, spleen and bone marrow (Fig. 1b). Single cell Proteomic analysis (CyTOF) of spleen showed reduced expression of cell proliferation marker -ki67, c-myc, ERK and p38 proteins, and reduction in number of leukemic cells. Finally, this combination therapy resulted in reduced leukemia burden and extension of overall survival across all three aggressive NOTCH1-mutated T-ALL PDX models (p Disclosures Jabbour: Pfizer: Other: Advisory role, Research Funding; Genentech: Other: Advisory role, Research Funding; BMS: Other: Advisory role, Research Funding; Takeda: Other: Advisory role, Research Funding; Amgen: Other: Advisory role, Research Funding; Adaptive Biotechnologies: Other: Advisory role, Research Funding; AbbVie: Other: Advisory role, Research Funding. Teachey:Sobi: Consultancy; Amgen: Consultancy; Janssen: Consultancy; La Roche: Consultancy. Rezvani:Takeda: Other: Licensing agreement; GemoAb: Membership on an entity's Board of Directors or advisory committees; Adicet Bio: Membership on an entity's Board of Directors or advisory committees; Virogen: Membership on an entity's Board of Directors or advisory committees; Pharmacyclics: Other: Educational grant; Affimed: Other: Educational grant; Formula Pharma: Membership on an entity's Board of Directors or advisory committees. Andreeff:Daiichi-Sankyo; Jazz Pharmaceuticals; Celgene; Amgen; AstraZeneca; 6 Dimensions Capital: Consultancy; Daiichi-Sankyo; Breast Cancer Research Foundation; CPRIT; NIH/NCI; Amgen; AstraZeneca: Research Funding; Centre for Drug Research & Development; Cancer UK; NCI-CTEP; German Research Council; Leukemia Lymphoma Foundation (LLS); NCI-RDCRN (Rare Disease Clin Network); CLL Founcdation; BioLineRx; SentiBio; Aptose Biosciences, Inc: Membership on an entity's Board of Directors or advisory committees; Amgen: Research Funding. Lorenzi:Precision Pathways: Consultancy. Konopleva:Calithera: Research Funding; Kisoji: Consultancy; AbbVie: Consultancy, Research Funding; Sanofi: Research Funding; Genentech: Consultancy, Research Funding; F. Hoffmann La-Roche: Consultancy, Research Funding; Cellectis: Research Funding; Rafael Pharmaceutical: Research Funding; Eli Lilly: Research Funding; Reata Pharmaceutical Inc.;: Patents & Royalties: patents and royalties with patent US 7,795,305 B2 on CDDO-compounds and combination therapies, licensed to Reata Pharmaceutical; Agios: Research Funding; AstraZeneca: Research Funding; Ablynx: Research Funding; Forty-Seven: Consultancy, Research Funding; Amgen: Consultancy; Stemline Therapeutics: Consultancy, Research Funding; Ascentage: Research Funding.
- Published
- 2020
- Full Text
- View/download PDF
40. GATA3-Controlled Nucleosome Eviction Drives MYC Enhancer Activity in T-cell Development and Leukemia
- Author
-
Ramya Raviram, Clara Reglero, Francesco Gavino Brundu, Raul Rabadan, Pedro P. Rocha, Jose R. Cortes, Alexander Y. Yang, Daniel Herranz, S. Aidan Quinn, Jules P.P. Meijerink, Pablo Pérez-Durán, Valentina Cordo, Agnieszka A. Wendorff, Francesca Gianni, Silvia Alvarez, Marissa Rashkovan, Robert Albero, Adolfo A. Ferrando, Devya Gurung, Laura Belver, and Anisha J. Cooke
- Subjects
0301 basic medicine ,Regulation of gene expression ,Effector ,HEK 293 cells ,Biology ,Jurkat cells ,Chromatin ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Gene expression ,Nucleosome ,Enhancer - Abstract
Long-range enhancers govern the temporal and spatial control of gene expression; however, the mechanisms that regulate enhancer activity during normal and malignant development remain poorly understood. Here, we demonstrate a role for aberrant chromatin accessibility in the regulation of MYC expression in T-cell lymphoblastic leukemia (T-ALL). Central to this process, the NOTCH1-MYC enhancer (N-Me), a long-range T cell–specific MYC enhancer, shows dynamic changes in chromatin accessibility during T-cell specification and maturation and an aberrant high degree of chromatin accessibility in mouse and human T-ALL cells. Mechanistically, we demonstrate that GATA3-driven nucleosome eviction dynamically modulates N-Me enhancer activity and is strictly required for NOTCH1-induced T-ALL initiation and maintenance. These results directly implicate aberrant regulation of chromatin accessibility at oncogenic enhancers as a mechanism of leukemic transformation. Significance: MYC is a major effector of NOTCH1 oncogenic programs in T-ALL. Here, we show a major role for GATA3-mediated enhancer nucleosome eviction as a driver of MYC expression and leukemic transformation. These results support the role of aberrant chromatin accessibility and consequent oncogenic MYC enhancer activation in NOTCH1-induced T-ALL. This article is highlighted in the In This Issue feature, p. 1631
- Published
- 2019
- Full Text
- View/download PDF
41. A selective BCL-XL PROTAC degrader achieves safe and potent antitumor activity
- Author
-
Yaxia Yuan, Yong-Mi Kim, Xingui Liu, Sajid Khan, Natalia Baran, Xuan Zhang, Yonghan He, Dinesh Thummuri, Weizhou Zhang, Peiyi Zhang, Janet S. Wiegand, Peter J. Houghton, Dongwen Lv, Vinitha Mary Kuruvilla, Christopher R. McCurdy, Guangcun Huang, Marina Konopleva, Robert Hromas, Guangrong Zheng, Qi Zhang, Anna Rogojina, Abhisheak Sharma, Jing Pei, Daohong Zhou, and Adolfo A. Ferrando
- Subjects
0301 basic medicine ,Navitoclax ,biology ,Chemistry ,Drug discovery ,Bcl-xL ,General Medicine ,medicine.disease ,General Biochemistry, Genetics and Molecular Biology ,In vitro ,03 medical and health sciences ,Leukemia ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,In vivo ,030220 oncology & carcinogenesis ,Cancer cell ,Toxicity ,biology.protein ,medicine ,Cancer research - Abstract
B-cell lymphoma extra large (BCL-XL) is a well-validated cancer target. However, the on-target and dose-limiting thrombocytopenia limits the use of BCL-XL inhibitors, such as ABT263, as safe and effective anticancer agents. To reduce the toxicity of ABT263, we converted it into DT2216, a BCL-XL proteolysis-targeting chimera (PROTAC), that targets BCL-XL to the Von Hippel-Lindau (VHL) E3 ligase for degradation. We found that DT2216 was more potent against various BCL-XL-dependent leukemia and cancer cells but considerably less toxic to platelets than ABT263 in vitro because VHL is poorly expressed in platelets. In vivo, DT2216 effectively inhibits the growth of several xenograft tumors as a single agent or in combination with other chemotherapeutic agents, without causing appreciable thrombocytopenia. These findings demonstrate the potential to use PROTAC technology to reduce on-target drug toxicities and rescue the therapeutic potential of previously undruggable targets. Furthermore, DT2216 may be developed as a safe first-in-class anticancer agent targeting BCL-XL. The first BCL-XL-degrading PROTAC achieves safer and more potent antitumor activity than dual BCL-XL and BCL-2 inhibitor navitoclax because of reduced dose-limiting platelet toxicity and high target specificity.
- Published
- 2019
- Full Text
- View/download PDF
42. Metabolic dependencies and vulnerabilities in leukemia
- Author
-
Adolfo A. Ferrando and Marissa Rashkovan
- Subjects
Anabolism ,medicine.medical_treatment ,Review ,Biology ,Mitochondrion ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,medicine ,Humans ,Cell Proliferation ,030304 developmental biology ,0303 health sciences ,Leukemia ,Cell growth ,medicine.disease ,Oncogene Addiction ,Warburg effect ,Metabolic pathway ,030220 oncology & carcinogenesis ,Cancer research ,Oxidation-Reduction ,Metabolic Networks and Pathways ,Developmental Biology - Abstract
Leukemia cell proliferation requires up-regulation and rewiring of metabolic pathways to feed anabolic cell growth. Oncogenic drivers directly and indirectly regulate metabolic pathways, and aberrant metabolism is central not only for leukemia proliferation and survival, but also mediates oncogene addiction with significant implications for the development of targeted therapies. This review explores leukemia metabolic circuitries feeding anabolism, redox potential, and energy required for tumor propagation with an emphasis on emerging therapeutic opportunities.
- Published
- 2019
- Full Text
- View/download PDF
43. Genetics and mechanisms of NT5C2-driven chemotherapy resistance in relapsed ALL
- Author
-
Chelsea L. Dieck and Adolfo A. Ferrando
- Subjects
Nucleotidase activity ,Immunology ,Mutant ,Biology ,medicine.disease_cause ,Biochemistry ,Acute lymphocytic leukemia ,Nucleotidase ,medicine ,Humans ,5'-Nucleotidase ,Cell Proliferation ,BLOOD Spotlight ,Mutation ,Thiopurine methyltransferase ,Mercaptopurine ,Cell Biology ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Chemotherapy regimen ,Neoplasm Proteins ,Leukemia ,Drug Resistance, Neoplasm ,Cancer research ,biology.protein - Abstract
Mutations in the cytosolic 59 nucleotidase II (NT5C2) gene drive resistance to thiopurine chemotherapy in relapsed acute lymphoblastic leukemia (ALL). Mechanistically, NT5C2 mutant proteins have increased nucleotidase activity as a result of altered activating and autoregulatory switch-off mechanisms. Leukemia cells harboring activating NT5C2 mutations are chemo-resistant to 6-mercaptopurine (6-MP), yet show impaired proliferative and self-renewal capacity. Direct inhibition of NT5C2 or pharmacologic targeting of compensatory pathways active in NT5C2 mutant cells may antagonize the emergence of NT5C2 mutant clones driving resistance and relapse in ALL.
- Published
- 2019
- Full Text
- View/download PDF
44. JAK-STAT inhibition mediates romidepsin and mechlorethamine synergism in Cutaneous T-cell Lymphoma
- Author
-
Teresa Palomero, Megan H. Trager, Jose R. Cortes, Anisha J. Cooke, Yuhan Gu, Marta Sanchez-Martin, Adam Mackey, Larisa J. Geskin, Anouchka P. Laurent, Adolfo A. Ferrando, Christina C. Patrone, Stuart Aidan Quinn, and Bobby B. Shih
- Subjects
0301 basic medicine ,Skin Neoplasms ,medicine.drug_class ,Dermatology ,Biochemistry ,Article ,Romidepsin ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Depsipeptides ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Janus Kinase Inhibitors ,Mechlorethamine ,Molecular Biology ,STAT5 ,biology ,business.industry ,Cutaneous T-cell lymphoma ,Histone deacetylase inhibitor ,JAK-STAT signaling pathway ,Drug Synergism ,Cell Biology ,medicine.disease ,Lymphoma ,Lymphoma, T-Cell, Cutaneous ,STAT Transcription Factors ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,STAT protein ,biology.protein ,Histone deacetylase ,business ,medicine.drug ,Signal Transduction - Abstract
Sezary syndrome is an aggressive and disseminated form of cutaneous T-cell lymphoma associated with dismal prognosis in which the histone deacetylase inhibitor romidepsin has shown remarkable activity as a single agent. However, clinical responses to romidepsin are typically transient, highlighting the need for more effective therapies. In this study, we show synergistic antilymphoma effects of romidepsin in combination with mechlorethamine, an alkylating agent, in cutaneous T-cell lymphoma cell lines and primary samples with strong antitumor effects in an in vivo model of Sezary syndrome. Mechanistically, gene expression profiling points to abrogation of Jak/signal transducer and activator of transcription (STAT) signaling as an important mediator of this interaction. Consistently, the combination of mechlorethamine plus romidepsin resulted in downregulation of STAT5 phosphorylation in romidepsin-sensitive cell lines and primary Sezary syndrome samples, but not in romidepsin-resistant tumors. Moreover, in further support of Jak/STAT signaling as a modulator of romidepsin activity in cutaneous T-cell lymphoma, treatment with romidepsin in combination with Jak inhibitors resulted in markedly increased therapeutic responses. Overall, these results support a role for romidepsin plus mechlorethamine in combination in the treatment of cutaneous T-cell lymphoma and uncover a previously unrecognized role for Jak/STAT signaling in the response to romidepsin and romidepsin-based combination therapies in Sezary syndrome.
- Published
- 2021
45. Tcf1 is essential for initiation of oncogenic Notch1-driven chromatin topology in T-ALL
- Author
-
Mateusz Antoszewski, Nadine Fournier, Gustavo A. Ruiz Buendía, Joao Lourenco, Yuanlong Liu, Tara Sugrue, Christelle Dubey, Marianne Nkosi, Colin E. J. Pritchard, Ivo J. Huijbers, Gabriela C. Segat, Sandra Alonso-Moreno, Elisabeth Serracanta, Laura Belver, Adolfo A. Ferrando, Giovanni Ciriello, Andrew P. Weng, Ute Koch, and Freddy Radtke
- Subjects
long-term ,Carcinogenesis ,Immunology ,beta-catenin ,Cell Biology ,Hematology ,Oncogenes ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Biochemistry ,gene-expression ,hematopoiesis ,Chromatin ,stem-cells ,specification ,cell development ,hemic and lymphatic diseases ,transcription factors ,Cell Line, Tumor ,Humans ,Receptor, Notch1 ,genome ,notch - Abstract
NOTCH1 is a well-established lineage specifier for T cells and among the most frequently mutated genes throughout all subclasses of T cell acute lymphoblastic leukemia (T-ALL). How oncogenic NOTCH1 signaling launches a leukemia-prone chromatin landscape during T-ALL initiation is unknown. Here we demonstrate an essential role for the high-mobility-group transcription factor Tcf1 in orchestrating chromatin accessibility and topology, allowing aberrant Notch1 signaling to convey its oncogenic function. Although essential, Tcf1 is not sufficient to initiate leukemia. The formation of a leukemia-prone epigenetic landscape at the distal Notch1-regulated Myc enhancer, which is fundamental to this disease, is Tcf1-dependent and occurs within the earliest progenitor stage even before cells adopt a T lymphocyte or leukemic fate. Moreover, we discovered a unique evolutionarily conserved Tcf1-regulated enhancer element in the distal Myc-enhancer, which is important for the transition of preleukemic cells to full-blown disease.
- Published
- 2021
46. Intracellular Cholesterol Pools Regulate Oncogenic Signaling and Epigenetic Circuitries in Early T-cell Precursor Acute Lymphoblastic Leukemia
- Author
-
Peter H. Wiernik, Mark R. Litzow, Maria Luisa Sulis, Pablo Perez-Duran, Francesca Gianni, Adolfo A. Ferrando, Selina M. Luger, Marissa Rashkovan, Jacob M. Rowe, Martin S. Tallman, Robert Albero, Hannah I Miller, Rajesh K Soni, Adam L Mackey, and Elisabeth Paietta
- Subjects
Precursor Cells, T-Lymphoid ,Cell growth ,Carcinogenesis ,T cell ,Mevalonic Acid ,Biology ,medicine.disease ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Article ,Chromatin ,Epigenesis, Genetic ,Transcriptome ,Proto-Oncogene Proteins c-myc ,Leukemia ,medicine.anatomical_structure ,Cholesterol ,Oncology ,medicine ,Cancer research ,Humans ,Mevalonate pathway ,Epigenetics ,Signal transduction ,Signal Transduction - Abstract
Early T-cell acute lymphoblastic leukemia (ETP-ALL) is an aggressive hematologic malignancy associated with early relapse and poor prognosis that is genetically, immunophenotypically, and transcriptionally distinct from more mature T-cell acute lymphoblastic leukemia (T-ALL) tumors. Here, we leveraged global metabolomic and transcriptomic profiling of primary ETP- and T-ALL leukemia samples to identify specific metabolic circuitries differentially active in this high-risk leukemia group. ETP-ALLs showed increased biosynthesis of phospholipids and sphingolipids and were specifically sensitive to inhibition of 3-hydroxy-3-methylglutaryl-CoA reductase, the rate-limiting enzyme in the mevalonate pathway. Mechanistically, inhibition of cholesterol synthesis inhibited oncogenic AKT1 signaling and suppressed MYC expression via loss of chromatin accessibility at a leukemia stem cell–specific long-range MYC enhancer. In all, these results identify the mevalonate pathway as a druggable novel vulnerability in high-risk ETP-ALL cells and uncover an unanticipated critical role for cholesterol biosynthesis in signal transduction and epigenetic circuitries driving leukemia cell growth and survival. Significance: Overtly distinct cell metabolic pathways operate in ETP- and T-ALL pointing to specific metabolic vulnerabilities. Inhibition of mevalonate biosynthesis selectively blocks oncogenic AKT–MYC signaling in ETP-ALL and suppresses leukemia cell growth. Ultimately, these results will inform the development of novel tailored and more effective treatments for patients with high-risk ETP-ALL. This article is highlighted in the In This Issue feature, p. 587
- Published
- 2021
47. Mutational and functional genetics mapping of chemotherapy resistance mechanisms in relapsed acute lymphoblastic leukemia
- Author
-
Martin S. Tallman, Junfei Zhao, Mignon L. Loh, Giuseppe Basso, Motohiro Kato, Meenakshi Devidas, Pablo Pérez-Durán, Timothy Chu, Julie M. Gastier-Foster, Maddalena Paganin, Alberto Ambesi-Impiombato, Katsuyoshi Koh, Zhengqiang Wang, Adolfo A. Ferrando, Laura Belver, Mark R. Litzow, Jessie A. Brown, Raul Rabadan, Concepcion Nicolas, Jules P.P. Meijerink, Elisabeth Paietta, Thomas Gunning, Aidan Quinn, Maria Luisa Sulis, Valeria Tosello, Juan Ángel Patiño-Galindo, Mark D. Minden, Jacob M. Rowe, Koichi Oshima, and Milagros Balbín
- Subjects
Drug ,Oncology ,Adult ,Cancer Research ,medicine.medical_specialty ,Lymphoblastic Leukemia ,media_common.quotation_subject ,Article ,03 medical and health sciences ,0302 clinical medicine ,Recurrence ,Internal medicine ,Acute lymphocytic leukemia ,Medicine ,CRISPR ,Humans ,Child ,media_common ,business.industry ,Cancer ,Combination chemotherapy ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Prognosis ,Leukemia ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Mutation ,business ,Chemotherapy resistance - Abstract
Multiagent combination chemotherapy can be curative in acute lymphoblastic leukemia (ALL). Still, patients with primary refractory disease or with relapsed leukemia have a very poor prognosis. Here we integrate an in-depth dissection of the mutational landscape across diagnostic and relapsed pediatric and adult ALL samples with genome-wide CRISPR screen analysis of gene–drug interactions across seven ALL chemotherapy drugs. By combining these analyses, we uncover diagnostic and relapse-specific mutational mechanisms as well as genetic drivers of chemoresistance. Functionally, our data identify common and drug-specific pathways modulating chemotherapy response and underscore the effect of drug combinations in restricting the selection of resistance-driving genetic lesions. In addition, by identifying actionable targets for the reversal of chemotherapy resistance, these analyses open therapeutic opportunities for the treatment of relapse and refractory disease. Ferrando and colleagues analyze matched diagnostic and relapsed acute lymphocytic leukemia by whole-genome sequencing, and perform in vitro genome-wide CRISPR screens, to examine alterations associated with chemotherapy resistance.
- Published
- 2021
48. Enhancer Hijacking Drives Oncogenic
- Author
-
Lindsey E, Montefiori, Sonja, Bendig, Zhaohui, Gu, Xiaolong, Chen, Petri, Pölönen, Xiaotu, Ma, Alex, Murison, Andy, Zeng, Laura, Garcia-Prat, Kirsten, Dickerson, Ilaria, Iacobucci, Sherif, Abdelhamed, Ryan, Hiltenbrand, Paul E, Mead, Cyrus M, Mehr, Beisi, Xu, Zhongshan, Cheng, Ti-Cheng, Chang, Tamara, Westover, Jing, Ma, Anna, Stengel, Shunsuke, Kimura, Chunxu, Qu, Marcus B, Valentine, Marissa, Rashkovan, Selina, Luger, Mark R, Litzow, Jacob M, Rowe, Monique L, den Boer, Victoria, Wang, Jun, Yin, Steven M, Kornblau, Stephen P, Hunger, Mignon L, Loh, Ching-Hon, Pui, Wenjian, Yang, Kristine R, Crews, Kathryn G, Roberts, Jun J, Yang, Mary V, Relling, William E, Evans, Wendy, Stock, Elisabeth M, Paietta, Adolfo A, Ferrando, Jinghui, Zhang, Wolfgang, Kern, Torsten, Haferlach, Gang, Wu, John E, Dick, Jeffery M, Klco, Claudia, Haferlach, and Charles G, Mullighan
- Subjects
Repressor Proteins ,Leukemia, Myeloid, Acute ,Enhancer Elements, Genetic ,Tumor Suppressor Proteins ,Humans ,Gene Regulatory Networks ,Hematopoietic Stem Cells ,Article ,Transcription Factors - Abstract
Lineage ambiguous leukemias are high-risk malignancies of poorly understood genetic basis. Here, we describe a distinct subgroup of acute leukemia with expression of myeloid, T lymphoid and stem cell markers driven by aberrant allele-specific deregulation of BCL11B, a master transcription factor responsible for thymic T-lineage commitment and specification. Mechanistically, this deregulation was driven by chromosomal rearrangements that juxtapose BCL11B to super-enhancers active in hematopoietic progenitors, or focal amplifications that generate a super-enhancer from a non-coding element distal to BCL11B. Chromatin conformation analyses demonstrate long range interactions of rearranged enhancers with the expressed BCL11B allele, and association of BCL11B with activated hematopoietic progenitor cell cis-regulatory elements, suggesting BCL11B is aberrantly co-opted into a gene regulatory network that drives transformation by maintaining a progenitor state. These data support a role for ectopic BCL11B expression in primitive hematopoietic cells mediated by enhancer hijacking as an oncogenic driver of human lineage ambiguous leukemia.
- Published
- 2021
49. Deregulation of enhancer structure, function, and dynamics in acute lymphoblastic leukemia
- Author
-
Robert Albero, Adolfo A. Ferrando, and Laura Belver
- Subjects
0301 basic medicine ,Oncogene ,Cell growth ,Carcinogenesis ,Lymphoblastic Leukemia ,Immunology ,Cell Differentiation ,Biology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Article ,Malignant transformation ,Cell biology ,03 medical and health sciences ,Mice ,030104 developmental biology ,0302 clinical medicine ,Enhancer Elements, Genetic ,Gene expression ,Immunology and Allergy ,Animals ,Epigenetics ,Enhancer ,Function (biology) ,030215 immunology - Abstract
Enhancers control dynamic changes in gene expression and orchestrate the tightly controlled transcriptional circuitries that direct and coordinate cell growth, proliferation, survival, lineage commitment, and differentiation during lymphoid development. Enhancer hijacking and neoenhancer formation at oncogene loci, as well as aberrant activation of oncogene-associated enhancers, can induce constitutive activation of self-perpetuating oncogenic transcriptional circuitries, and contribute to the malignant transformation of immature lymphoid progenitors in acute lymphoblastic leukemia (ALL). In this review, we present recent discoveries on the role of enhancer dynamics in mouse and human lymphoid development, discuss how genetic and epigenetic alterations of enhancer function can promote leukemogenesis, and discuss potential strategies for targeting the enhancer machinery in the treatment of ALL.
- Published
- 2021
50. FYN-TRAF3IP2 induces NF-κB signaling-driven peripheral T cell lymphoma
- Author
-
Raul Rabadan, Jose R. Cortes, Adolfo A. Ferrando, Craig R. Soderquist, Teresa Palomero, Anisha J. Cooke, Clara Reglero, Silvia Alvarez, Junfei Zhao, Elias Campo, Christine S. Moon, S. Aidan Quinn, Govind Bhagat, Claudia Fiñana, Francesco Abate, Wen-Hsuan Lin, and Giorgio Inghirami
- Subjects
Cancer Research ,Angioimmunoblastic T-cell lymphoma ,business.industry ,T-Cell Transformation ,IκB kinase ,medicine.disease ,T-Cell Receptor Activation ,Peripheral T-cell lymphoma ,Article ,Haematopoiesis ,FYN ,Oncology ,medicine ,Cancer research ,Progenitor cell ,business - Abstract
Angioimmunoblastic T cell lymphoma (AITL) and peripheral T cell lymphoma not-otherwise-specified (PTCL, NOS) have poor prognosis and lack driver actionable targets for directed therapies in most cases. Here we identify FYN-TRAF3IP2 as a recurrent oncogenic gene fusion in AITL and PTCL, NOS tumors. Mechanistically, we show that FYN-TRAF3IP2 leads to aberrant NF-κB signaling downstream of T cell receptor activation. Consistent with a driver oncogenic role, FYN-TRAF3IP2 expression in hematopoietic progenitors induces NF-κB-driven T cell transformation in mice and cooperates with loss of the Tet2 tumor suppressor in PTCL development. Moreover, abrogation of NF-κB signaling in FYN-TRAF3IP2-induced tumors with IκB kinase inhibitors delivers strong anti-lymphoma effects in vitro and in vivo. These results demonstrate an oncogenic and pharmacologically targetable role for FYN-TRAF3IP2 in PTCLs and call for the clinical testing of anti-NF-κB targeted therapies in these diseases.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.