138 results on '"Alexandre Puissant"'
Search Results
2. A multiparametric niche-like drug screening platform in acute myeloid leukemia
- Author
-
Reinaldo Dal Bello, Justine Pasanisi, Romane Joudinaud, Matthieu Duchmann, Bryann Pardieu, Paolo Ayaka, Giuseppe Di Feo, Gaetano Sodaro, Clémentine Chauvel, Rathana Kim, Loic Vasseur, Laureen Chat, Frank Ling, Kim Pacchiardi, Camille Vaganay, Jeannig Berrou, Chaima Benaksas, Nicolas Boissel, Thorsten Braun, Claude Preudhomme, Hervé Dombret, Emmanuel Raffoux, Nina Fenouille, Emmanuelle Clappier, Lionel Adès, Alexandre Puissant, and Raphael Itzykson
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Functional precision medicine in AML often relies on short-term in vitro drug sensitivity screening (DSS) of primary patient cells in standard culture conditions. We designed a niche-like DSS assay combining physiologic hypoxia (O2 3%) and mesenchymal stromal cell (MSC) co-culture with multiparameter flow cytometry to enumerate lymphocytes and differentiating (CD11/CD14/CD15+) or leukemic stem cell (LSC)-enriched (GPR56+) cells within the leukemic bulk. After functional validation of GPR56 expression as a surrogate for LSC enrichment, the assay identified three patterns of response, including cytotoxicity on blasts sparing LSCs, induction of differentiation, and selective impairment of LSCs. We refined our niche-like culture by including plasma-like amino-acid and cytokine concentrations identified by targeted metabolomics and proteomics of primary AML bone marrow plasma samples. Systematic interrogation revealed distinct contributions of each niche-like component to leukemic outgrowth and drug response. Short-term niche-like culture preserved clonal architecture and transcriptional states of primary leukemic cells. In a cohort of 45 AML samples enriched for NPM1c AML, the niche-like multiparametric assay could predict morphologically (p = 0.02) and molecular (NPM1c MRD, p = 0.04) response to anthracycline-cytarabine induction chemotherapy. In this cohort, a 23-drug screen nominated ruxolitinib as a sensitizer to anthracycline-cytarabine. This finding was validated in an NPM1c PDX model.
- Published
- 2022
- Full Text
- View/download PDF
3. Favorable pharmacokinetic and pharmacodynamic properties of gilteritinib in cerebrospinal fluid: a potential effective treatment in relapsing meningeal acute myeloid leukemia FLT3-ITD patients
- Author
-
Nicolas Vignal, Loïs Kelly, Etienne Lengline, Aurélie Cabannes-Hamy, Justine Siavellis, David Ghez, Hélène Sauvageon, Thorsten Braun, Evelyne Jacqz-Aigrain, Milena Kohn, Philippe Rousselot, Alexandre Puissant, Emmanuel Raffoux, Samia Mourah, and Lauriane Goldwirt
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
4. Relative Mitochondrial Priming Predicts Survival in Older AML Patients Treated Intensively
- Author
-
Reinaldo Dal Bello, Kim Pacchiardi, Clémentine Chauvel, Lionel Adès, Thorsten Braun, Justine Pasanisi, Elise Fournier, Céline Berthon, Emmanuelle Clappier, Emmanuel Raffoux, Delphine Lebon, Thomas Cluzeau, Christophe Roumier, Adriana Plesa, Karine Celli-Lebras, Hervé Dombret, Claude Preudhomme, Stéphanie Mathis, Alexandre Puissant, Claude Gardin, and Raphael Itzykson
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
5. Acadesine kills chronic myelogenous leukemia (CML) cells through PKC-dependent induction of autophagic cell death.
- Author
-
Guillaume Robert, Issam Ben Sahra, Alexandre Puissant, Pascal Colosetti, Nathalie Belhacene, Pierre Gounon, Paul Hofman, Fréderic Bost, Jill-Patrice Cassuto, and Patrick Auberger
- Subjects
Medicine ,Science - Abstract
CML is an hematopoietic stem cell disease characterized by the t(9;22) (q34;q11) translocation encoding the oncoprotein p210BCR-ABL. The effect of acadesine (AICAR, 5-Aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside) a compound with known antileukemic effect on B cell chronic lymphoblastic leukemia (B-CLL) was investigated in different CML cell lines. Acadesine triggered loss of cell metabolism in K562, LAMA-84 and JURL-MK1 and was also effective in killing imatinib-resistant K562 cells and Ba/F3 cells carrying the T315I-BCR-ABL mutation. The anti-leukemic effect of acadesine did not involve apoptosis but required rather induction of autophagic cell death. AMPK knock-down by Sh-RNA failed to prevent the effect of acadesine, indicating an AMPK-independent mechanism. The effect of acadesine was abrogated by GF109203X and Ro-32-0432, both inhibitor of classical and new PKCs and accordingly, acadesine triggered relocation and activation of several PKC isoforms in K562 cells. In addition, this compound exhibited a potent anti-leukemic effect in clonogenic assays of CML cells in methyl cellulose and in a xenograft model of K562 cells in nude mice. In conclusion, our work identifies an original and unexpected mechanism by which acadesine triggers autophagic cell death through PKC activation. Therefore, in addition to its promising effects in B-CLL, acadesine might also be beneficial for Imatinib-resistant CML patients.
- Published
- 2009
- Full Text
- View/download PDF
6. Dual role of Sp3 transcription factor as an inducer of apoptosis and a marker of tumour aggressiveness.
- Author
-
Khadija Essafi-Benkhadir, Sébastien Grosso, Alexandre Puissant, Guillaume Robert, Makram Essafi, Marcel Deckert, Emmanuel Chamorey, Olivier Dassonville, Gérard Milano, Patrick Auberger, and Gilles Pagès
- Subjects
Medicine ,Science - Abstract
BackgroundThe ambiguous role of transcription factor Sp3 for tumour progression is still debated since it was described as a transcriptional repressor or activator. Here we tried to decipher the molecular mechanisms implicated in Sp3 accumulation observed in aggressive tumours.MethodologyWe generated normal and tumour cell lines conditionally expressing Sp3. Cell growth was analyzed in vitro and after inoculation in nude mice. Apoptosis was assessed by pan- caspase activity assays, by counting fragmented nuclei and by determination of caspase 9 cleavage. Gene expression was determined by quantitative PCR. Cleavage by different caspases was performed after in vitro translation of the Sp3 cDNA in the presence of [S(35)] labelled methionine. Different tumour cell lines and head and neck tumour samples were tested for the presence of Sp3 by western blots. Correlation between Sp3 expression and overall survival has been statistically determined.Principal findingsConditional over-expression of Sp3 induces apoptosis and modifies expression of genes implicated in the regulation of cell cycle and pro and anti apoptotic genes. Sp3 over-expression strongly reduces the development of tumours in nude mice confirming its pro-apoptotic potential in vivo. However, cells can survive to apoptosis through selective Sp3 cleavage by caspase. Sp3 induction in established tumours resulted in transient regression then progression. Progression coincides with re-accumulation of the full length form of Sp3. Sp3 is over-expressed in tumour cell lines of different origins. The presence of high levels of the full-length form of Sp3 indicates a poor prognosis for overall survival of patients with head and neck tumours.ConclusionsFull length Sp3 accumulation highlights bypass of tumour cell apoptotic capacities and is indicative of head and neck tumours aggressiveness.
- Published
- 2009
- Full Text
- View/download PDF
7. P2RY2-AKT activation is a therapeutically actionable consequence of XPO1 inhibition in acute myeloid leukemia
- Author
-
Kevin H. Lin, Justine C. Rutter, Abigail Xie, Shane T. Killarney, Camille Vaganay, Chaima Benaksas, Frank Ling, Gaetano Sodaro, Paul-Arthur Meslin, Christopher F. Bassil, Nina Fenouille, Jacob Hoj, Rachel Washart, Hazel X. Ang, Christian Cerda-Smith, Paul Chaintreuil, Arnaud Jacquel, Patrick Auberger, Antoine Forget, Raphael Itzykson, Min Lu, Jiaxing Lin, Mariaelena Pierobon, Zhecheng Sheng, Xinghai Li, Ashutosh Chilkoti, Kouros Owzar, David A. Rizzieri, Timothy S. Pardee, Lina Benajiba, Emanuel Petricoin, Alexandre Puissant, and Kris C. Wood
- Subjects
Receptors, Purinergic P2Y2 ,Leukemia, Myeloid, Acute ,Mice ,Cancer Research ,Oncology ,Antineoplastic Combined Chemotherapy Protocols ,Animals ,Receptors, Cytoplasmic and Nuclear ,Karyopherins ,Proto-Oncogene Proteins c-akt ,United States - Abstract
Selinexor is a first-in-class inhibitor of the nuclear exportin XPO1 that was recently approved by the US Food and Drug Administration for the treatment of multiple myeloma and diffuse large B-cell lymphoma. In relapsed/refractory acute myeloid leukemia (AML), selinexor has shown promising activity, suggesting that selinexor-based combination therapies may have clinical potential. Here, motivated by the hypothesis that selinexor's nuclear sequestration of diverse substrates imposes pleiotropic fitness effects on AML cells, we systematically catalog the pro- and anti-fitness consequences of selinexor treatment. We discover that selinexor activates PI3Kγ-dependent AKT signaling in AML by upregulating the purinergic receptor P2RY2. Inhibiting this axis potentiates the anti-leukemic effects of selinexor in AML cell lines, patient-derived primary cultures and multiple mouse models of AML. In a syngeneic, MLL-AF9-driven mouse model of AML, treatment with selinexor and ipatasertib outperforms both standard-of-care chemotherapy and chemotherapy with selinexor. Together, these findings establish drug-induced P2RY2-AKT signaling as an actionable consequence of XPO1 inhibition in AML.
- Published
- 2022
- Full Text
- View/download PDF
8. Figure S10 from The Folate Cycle Enzyme MTHFR Is a Critical Regulator of Cell Response to MYC-Targeting Therapies
- Author
-
Alexandre Puissant, Raphael Itzykson, Kris C. Wood, Camille Lobry, Kimberly Stegmaier, Hava M. Golan, Lina Benajiba, Nina Fenouille, Claude Preudhomme, Hervé Dombret, Thomas Cluzeau, Giovanni Roti, Jean-Baptiste Micol, Marie-Hélène Schlageter, Jun Qi, Yana Pikman, Alain Pruvost, Jean-François Benoist, Gabriela Alexe, Iléana Antony-Debré, Justine Pasanisi, Gael Fortin, Christopher F. Bassil, Justine C. Rutter, Kevin H. Lin, Bryann Pardieu, Antoine Forget, Reinaldo Dal Bello, Chaïma Benaksas, Gaetano Sodaro, Camille Vaganay, Frank Ling, and Angela Su
- Abstract
Metabolic perturbations induced by folate starvation in AML cells.
- Published
- 2023
- Full Text
- View/download PDF
9. Figure S1 from Matched Targeted Therapy for Pediatric Patients with Relapsed, Refractory, or High-Risk Leukemias: A Report from the LEAP Consortium
- Author
-
Kimberly Stegmaier, Mignon L. Loh, Marian H. Harris, Jeffrey W. Tyner, Lewis B. Silverman, Katherine A. Janeway, Lia Gore, Neal I. Lindeman, Annette S. Kim, Stephen P. Hunger, Alexandre Puissant, Andrew E. Place, Elliot Stieglitz, Katherine Tarlock, Amy Saur Conway, Amanda L. Robichaud, Angela Su, Neekesh V. Dharia, Matthew P. Jacobson, Diego Garrido Ruiz, Wilian A. Cortopassi, Anjali Cremer, Cristina F. Contreras, Haley L. Faust, Tasleema Patel, Cristina E. Tognon, Alma Imamovic, Shan Lin, Yuting Li, Giacomo Gotti, Nicole Ocasio-Martinez, Catherine M. Clinton, Asmani A. Adhav, Lisa Gennarini, Jeffrey A. Magee, Matthew J. Barth, Jing Chen, Justine M. Kahn, Peter D. Cole, Neerav N. Shukla, Jennifer L. McNeer, Nathan Gossai, Patrick A. Brown, Michael J. Burke, Kelly W. Maloney, Melinda Pauly, Todd M. Cooper, Beth Apsel Winger, Traci M. Blonquist, Kristen Stevenson, Maria Luisa Sulis, Sarah K. Tasian, and Yana Pikman
- Abstract
VAF concordance
- Published
- 2023
- Full Text
- View/download PDF
10. Table S6 from The Folate Cycle Enzyme MTHFR Is a Critical Regulator of Cell Response to MYC-Targeting Therapies
- Author
-
Alexandre Puissant, Raphael Itzykson, Kris C. Wood, Camille Lobry, Kimberly Stegmaier, Hava M. Golan, Lina Benajiba, Nina Fenouille, Claude Preudhomme, Hervé Dombret, Thomas Cluzeau, Giovanni Roti, Jean-Baptiste Micol, Marie-Hélène Schlageter, Jun Qi, Yana Pikman, Alain Pruvost, Jean-François Benoist, Gabriela Alexe, Iléana Antony-Debré, Justine Pasanisi, Gael Fortin, Christopher F. Bassil, Justine C. Rutter, Kevin H. Lin, Bryann Pardieu, Antoine Forget, Reinaldo Dal Bello, Chaïma Benaksas, Gaetano Sodaro, Camille Vaganay, Frank Ling, and Angela Su
- Abstract
List of sgRNA Guides Used in the CRISPR Screen.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Methods from Matched Targeted Therapy for Pediatric Patients with Relapsed, Refractory, or High-Risk Leukemias: A Report from the LEAP Consortium
- Author
-
Kimberly Stegmaier, Mignon L. Loh, Marian H. Harris, Jeffrey W. Tyner, Lewis B. Silverman, Katherine A. Janeway, Lia Gore, Neal I. Lindeman, Annette S. Kim, Stephen P. Hunger, Alexandre Puissant, Andrew E. Place, Elliot Stieglitz, Katherine Tarlock, Amy Saur Conway, Amanda L. Robichaud, Angela Su, Neekesh V. Dharia, Matthew P. Jacobson, Diego Garrido Ruiz, Wilian A. Cortopassi, Anjali Cremer, Cristina F. Contreras, Haley L. Faust, Tasleema Patel, Cristina E. Tognon, Alma Imamovic, Shan Lin, Yuting Li, Giacomo Gotti, Nicole Ocasio-Martinez, Catherine M. Clinton, Asmani A. Adhav, Lisa Gennarini, Jeffrey A. Magee, Matthew J. Barth, Jing Chen, Justine M. Kahn, Peter D. Cole, Neerav N. Shukla, Jennifer L. McNeer, Nathan Gossai, Patrick A. Brown, Michael J. Burke, Kelly W. Maloney, Melinda Pauly, Todd M. Cooper, Beth Apsel Winger, Traci M. Blonquist, Kristen Stevenson, Maria Luisa Sulis, Sarah K. Tasian, and Yana Pikman
- Abstract
Supplementary Data file including supplementary methods and clinical case vignettes.
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Tables 1 - 9 from Targeting MYCN in Neuroblastoma by BET Bromodomain Inhibition
- Author
-
Kimberly Stegmaier, James E. Bradner, William A. Weiss, Andrew L. Kung, Cyril H. Benes, Ultan McDermott, Matthew J. Garnett, Patricia Greninger, William Clay Gustafson, Rhamy Zeid, Erin A. Nekritz, Yvan H. Chanthery, Jun Qi, Christopher F. Bassil, Gabriela Alexe, Stacey M. Frumm, and Alexandre Puissant
- Abstract
PDF file - 93K, This file contains 9 supplementary tables including the full primary screening data and secondary testing of BETi in neuroblastoma, supporting tables for the expression profiling analyses, and the list of Affymetrix probes (MYCN) and shRNA sequences for BRD4 and MYCN.
- Published
- 2023
- Full Text
- View/download PDF
13. Table S3 from Matched Targeted Therapy for Pediatric Patients with Relapsed, Refractory, or High-Risk Leukemias: A Report from the LEAP Consortium
- Author
-
Kimberly Stegmaier, Mignon L. Loh, Marian H. Harris, Jeffrey W. Tyner, Lewis B. Silverman, Katherine A. Janeway, Lia Gore, Neal I. Lindeman, Annette S. Kim, Stephen P. Hunger, Alexandre Puissant, Andrew E. Place, Elliot Stieglitz, Katherine Tarlock, Amy Saur Conway, Amanda L. Robichaud, Angela Su, Neekesh V. Dharia, Matthew P. Jacobson, Diego Garrido Ruiz, Wilian A. Cortopassi, Anjali Cremer, Cristina F. Contreras, Haley L. Faust, Tasleema Patel, Cristina E. Tognon, Alma Imamovic, Shan Lin, Yuting Li, Giacomo Gotti, Nicole Ocasio-Martinez, Catherine M. Clinton, Asmani A. Adhav, Lisa Gennarini, Jeffrey A. Magee, Matthew J. Barth, Jing Chen, Justine M. Kahn, Peter D. Cole, Neerav N. Shukla, Jennifer L. McNeer, Nathan Gossai, Patrick A. Brown, Michael J. Burke, Kelly W. Maloney, Melinda Pauly, Todd M. Cooper, Beth Apsel Winger, Traci M. Blonquist, Kristen Stevenson, Maria Luisa Sulis, Sarah K. Tasian, and Yana Pikman
- Abstract
Reasons for not using recommended targeted therapy.
- Published
- 2023
- Full Text
- View/download PDF
14. Data from The Folate Cycle Enzyme MTHFR Is a Critical Regulator of Cell Response to MYC-Targeting Therapies
- Author
-
Alexandre Puissant, Raphael Itzykson, Kris C. Wood, Camille Lobry, Kimberly Stegmaier, Hava M. Golan, Lina Benajiba, Nina Fenouille, Claude Preudhomme, Hervé Dombret, Thomas Cluzeau, Giovanni Roti, Jean-Baptiste Micol, Marie-Hélène Schlageter, Jun Qi, Yana Pikman, Alain Pruvost, Jean-François Benoist, Gabriela Alexe, Iléana Antony-Debré, Justine Pasanisi, Gael Fortin, Christopher F. Bassil, Justine C. Rutter, Kevin H. Lin, Bryann Pardieu, Antoine Forget, Reinaldo Dal Bello, Chaïma Benaksas, Gaetano Sodaro, Camille Vaganay, Frank Ling, and Angela Su
- Abstract
Deciphering the impact of metabolic intervention on response to anticancer therapy may elucidate a path toward improved clinical responses. Here, we identify amino acid–related pathways connected to the folate cycle whose activation predicts sensitivity to MYC-targeting therapies in acute myeloid leukemia (AML). We establish that folate restriction and deficiency of the rate-limiting folate cycle enzyme MTHFR, which exhibits reduced-function polymorphisms in about 10% of Caucasians, induce resistance to MYC targeting by BET and CDK7 inhibitors in cell lines, primary patient samples, and syngeneic mouse models of AML. Furthermore, this effect is abrogated by supplementation with the MTHFR enzymatic product CH3-THF. Mechanistically, folate cycle disturbance reduces H3K27/K9 histone methylation and activates a SPI1 transcriptional program counteracting the effect of BET inhibition. Our data provide a rationale for screening MTHFR polymorphisms and folate cycle status to nominate patients most likely to benefit from MYC-targeting therapies.Significance:Although MYC-targeting therapies represent a promising strategy for cancer treatment, evidence of predictors of sensitivity to these agents is limited. We pinpoint that folate cycle disturbance and frequent polymorphisms associated with reduced MTHFR activity promote resistance to BET inhibitors. CH3-THF supplementation thus represents a low-risk intervention to enhance their effects.See related commentary by Marando and Huntly, p. 1791.This article is highlighted in the In This Issue feature, p. 1775
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Figures 1 -11 from Targeting MYCN in Neuroblastoma by BET Bromodomain Inhibition
- Author
-
Kimberly Stegmaier, James E. Bradner, William A. Weiss, Andrew L. Kung, Cyril H. Benes, Ultan McDermott, Matthew J. Garnett, Patricia Greninger, William Clay Gustafson, Rhamy Zeid, Erin A. Nekritz, Yvan H. Chanthery, Jun Qi, Christopher F. Bassil, Gabriela Alexe, Stacey M. Frumm, and Alexandre Puissant
- Abstract
PDF file - 326K, This file contains 11 supplementary figures with biochemistry, computational analysis, in vitro and in vivo studies to support the role of targeting MYCN in neuroblastoma with BETi.
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Data from The Folate Cycle Enzyme MTHFR Is a Critical Regulator of Cell Response to MYC-Targeting Therapies
- Author
-
Alexandre Puissant, Raphael Itzykson, Kris C. Wood, Camille Lobry, Kimberly Stegmaier, Hava M. Golan, Lina Benajiba, Nina Fenouille, Claude Preudhomme, Hervé Dombret, Thomas Cluzeau, Giovanni Roti, Jean-Baptiste Micol, Marie-Hélène Schlageter, Jun Qi, Yana Pikman, Alain Pruvost, Jean-François Benoist, Gabriela Alexe, Iléana Antony-Debré, Justine Pasanisi, Gael Fortin, Christopher F. Bassil, Justine C. Rutter, Kevin H. Lin, Bryann Pardieu, Antoine Forget, Reinaldo Dal Bello, Chaïma Benaksas, Gaetano Sodaro, Camille Vaganay, Frank Ling, and Angela Su
- Abstract
Supplementary Material and Methods
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Data from Matched Targeted Therapy for Pediatric Patients with Relapsed, Refractory, or High-Risk Leukemias: A Report from the LEAP Consortium
- Author
-
Kimberly Stegmaier, Mignon L. Loh, Marian H. Harris, Jeffrey W. Tyner, Lewis B. Silverman, Katherine A. Janeway, Lia Gore, Neal I. Lindeman, Annette S. Kim, Stephen P. Hunger, Alexandre Puissant, Andrew E. Place, Elliot Stieglitz, Katherine Tarlock, Amy Saur Conway, Amanda L. Robichaud, Angela Su, Neekesh V. Dharia, Matthew P. Jacobson, Diego Garrido Ruiz, Wilian A. Cortopassi, Anjali Cremer, Cristina F. Contreras, Haley L. Faust, Tasleema Patel, Cristina E. Tognon, Alma Imamovic, Shan Lin, Yuting Li, Giacomo Gotti, Nicole Ocasio-Martinez, Catherine M. Clinton, Asmani A. Adhav, Lisa Gennarini, Jeffrey A. Magee, Matthew J. Barth, Jing Chen, Justine M. Kahn, Peter D. Cole, Neerav N. Shukla, Jennifer L. McNeer, Nathan Gossai, Patrick A. Brown, Michael J. Burke, Kelly W. Maloney, Melinda Pauly, Todd M. Cooper, Beth Apsel Winger, Traci M. Blonquist, Kristen Stevenson, Maria Luisa Sulis, Sarah K. Tasian, and Yana Pikman
- Abstract
Patient sequencing data, by patient number and diagnosis.
- Published
- 2023
- Full Text
- View/download PDF
18. Data from Gene expression profiling of imatinib and PD166326-resistant CML cell lines identifies Fyn as a gene associated with resistance to BCR-ABL inhibitors
- Author
-
Patrick Auberger, Bernard Mari, Jill Patrice Cassuto, Marcel Deckert, Pascal Barbry, Kevin Lebrigand, Arnaud Jacquel, Pascal Colosetti, Maeva Dufies, Alexandre Puissant, and Sébastien Grosso
- Abstract
Imatinib is used to treat chronic myelogenous leukemia (CML), but resistance develops in all phases of this disease. The purpose of the present study was to identify the mode of resistance of newly derived imatinib-resistant (IM-R) and PD166326-resistant (PD-R) CML cells. IM-R and PD-R clones exhibited an increase in viability and a decrease in caspase activation in response to various doses of imatinib and PD166326, respectively, as compared with parental K562 cells. Resistance involved neither mutations in BCR-ABL nor increased BCR-ABL, MDR1 or Lyn expression, all known modes of resistance. To gain insight into the resistance mechanisms, we used pangenomic microarrays and identified 281 genes modulated in parental versus IM-R and PD-R cells. The gene signature was similar for IM-R and PD-R cells, accordingly with the cross-sensitivity observed for both inhibitors. These genes were functionally associated with pathways linked to development, cell adhesion, cell growth, and the JAK-STAT cascade. Especially relevant were the increased expression of the tyrosine kinases AXL and Fyn as well as CD44 and HMGA2. Small interfering RNA experiments and pharmacologic approaches identified FYN as a candidate for resistance to imatinib. Our findings provide a comprehensive picture of the transcriptional events associated with imatinib and PD166326 resistance and identify Fyn as a new potential target for therapeutic intervention in CML. [Mol Cancer Ther 2009;8(7):1924–33]
- Published
- 2023
- Full Text
- View/download PDF
19. Supplemental Figure 1 from Gene expression profiling of imatinib and PD166326-resistant CML cell lines identifies Fyn as a gene associated with resistance to BCR-ABL inhibitors
- Author
-
Patrick Auberger, Bernard Mari, Jill Patrice Cassuto, Marcel Deckert, Pascal Barbry, Kevin Lebrigand, Arnaud Jacquel, Pascal Colosetti, Maeva Dufies, Alexandre Puissant, and Sébastien Grosso
- Abstract
Legend to supplemental Figure 1: Ingenuity Pathway Analysis identifies a network of genes upregulated in both resistant clones compared to parental cells. The network is displayed graphically as nodes (genes/gene products) and edges (the biological relationships between the nodes). All grey nodes correspond to up-regulated genes in both IM-R and PD-R resistant cells. As described in the legend provided, nodes are displayed using various shapes that represent the functional class of the gene product. Edges are displayed with various labels that describe the nature of the relationship between the nodes (A, activation; B, binding; E, expression; I, inhibition; P, phosphorylation; T, transcription). Edges without a label represent binding only. White nodes were identified by the pathway analysis as part of the network.
- Published
- 2023
- Full Text
- View/download PDF
20. Supplemental Table 1 from Gene expression profiling of imatinib and PD166326-resistant CML cell lines identifies Fyn as a gene associated with resistance to BCR-ABL inhibitors
- Author
-
Patrick Auberger, Bernard Mari, Jill Patrice Cassuto, Marcel Deckert, Pascal Barbry, Kevin Lebrigand, Arnaud Jacquel, Pascal Colosetti, Maeva Dufies, Alexandre Puissant, and Sébastien Grosso
- Abstract
All significant (p {less than or equal to} 0.05) categories resulting from each list produced the over-represented biological themes shown in supplementary Table 1.
- Published
- 2023
- Full Text
- View/download PDF
21. Supplementary Methods from Persistent Activation of the Fyn/ERK Kinase Signaling Axis Mediates Imatinib Resistance in Chronic Myelogenous Leukemia Cells through Upregulation of Intracellular SPARC
- Author
-
Patrick Auberger, Sophie Tartare-Deckert, Sophie Raynaud, Jill-Patrice Cassuto, François-Xavier Mahon, Jean-Max Pasquet, Marcel Deckert, Mickaël Ohanna, Arnaud Jacquel, Guillaume Robert, Maeva Dufies, Alexandre Puissant, and Nina Fenouille
- Abstract
Supplementary Methods from Persistent Activation of the Fyn/ERK Kinase Signaling Axis Mediates Imatinib Resistance in Chronic Myelogenous Leukemia Cells through Upregulation of Intracellular SPARC
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary Figure 4 from Resveratrol Promotes Autophagic Cell Death in Chronic Myelogenous Leukemia Cells via JNK-Mediated p62/SQSTM1 Expression and AMPK Activation
- Author
-
Patrick Auberger, Sophie Raynaud, Jill-Patrice Cassuto, Frederic Luciano, Nina Fenouille, Guillaume Robert, and Alexandre Puissant
- Abstract
Supplementary Figure 4 from Resveratrol Promotes Autophagic Cell Death in Chronic Myelogenous Leukemia Cells via JNK-Mediated p62/SQSTM1 Expression and AMPK Activation
- Published
- 2023
- Full Text
- View/download PDF
23. Supplementary Figure Legends 1-4 from Resveratrol Promotes Autophagic Cell Death in Chronic Myelogenous Leukemia Cells via JNK-Mediated p62/SQSTM1 Expression and AMPK Activation
- Author
-
Patrick Auberger, Sophie Raynaud, Jill-Patrice Cassuto, Frederic Luciano, Nina Fenouille, Guillaume Robert, and Alexandre Puissant
- Abstract
Supplementary Figure Legends 1-4 from Resveratrol Promotes Autophagic Cell Death in Chronic Myelogenous Leukemia Cells via JNK-Mediated p62/SQSTM1 Expression and AMPK Activation
- Published
- 2023
- Full Text
- View/download PDF
24. Supplementary Materials and Methods from Resveratrol Promotes Autophagic Cell Death in Chronic Myelogenous Leukemia Cells via JNK-Mediated p62/SQSTM1 Expression and AMPK Activation
- Author
-
Patrick Auberger, Sophie Raynaud, Jill-Patrice Cassuto, Frederic Luciano, Nina Fenouille, Guillaume Robert, and Alexandre Puissant
- Abstract
Supplementary Materials and Methods from Resveratrol Promotes Autophagic Cell Death in Chronic Myelogenous Leukemia Cells via JNK-Mediated p62/SQSTM1 Expression and AMPK Activation
- Published
- 2023
- Full Text
- View/download PDF
25. Supplementary Figure 1 from Resveratrol Promotes Autophagic Cell Death in Chronic Myelogenous Leukemia Cells via JNK-Mediated p62/SQSTM1 Expression and AMPK Activation
- Author
-
Patrick Auberger, Sophie Raynaud, Jill-Patrice Cassuto, Frederic Luciano, Nina Fenouille, Guillaume Robert, and Alexandre Puissant
- Abstract
Supplementary Figure 1 from Resveratrol Promotes Autophagic Cell Death in Chronic Myelogenous Leukemia Cells via JNK-Mediated p62/SQSTM1 Expression and AMPK Activation
- Published
- 2023
- Full Text
- View/download PDF
26. Supplementary Figure 2 from Resveratrol Promotes Autophagic Cell Death in Chronic Myelogenous Leukemia Cells via JNK-Mediated p62/SQSTM1 Expression and AMPK Activation
- Author
-
Patrick Auberger, Sophie Raynaud, Jill-Patrice Cassuto, Frederic Luciano, Nina Fenouille, Guillaume Robert, and Alexandre Puissant
- Abstract
Supplementary Figure 2 from Resveratrol Promotes Autophagic Cell Death in Chronic Myelogenous Leukemia Cells via JNK-Mediated p62/SQSTM1 Expression and AMPK Activation
- Published
- 2023
- Full Text
- View/download PDF
27. Supplementary Figure Legends 1-6 from Persistent Activation of the Fyn/ERK Kinase Signaling Axis Mediates Imatinib Resistance in Chronic Myelogenous Leukemia Cells through Upregulation of Intracellular SPARC
- Author
-
Patrick Auberger, Sophie Tartare-Deckert, Sophie Raynaud, Jill-Patrice Cassuto, François-Xavier Mahon, Jean-Max Pasquet, Marcel Deckert, Mickaël Ohanna, Arnaud Jacquel, Guillaume Robert, Maeva Dufies, Alexandre Puissant, and Nina Fenouille
- Abstract
Supplementary Figure Legends 1-6 from Persistent Activation of the Fyn/ERK Kinase Signaling Axis Mediates Imatinib Resistance in Chronic Myelogenous Leukemia Cells through Upregulation of Intracellular SPARC
- Published
- 2023
- Full Text
- View/download PDF
28. Supplementary Figure 3 from Resveratrol Promotes Autophagic Cell Death in Chronic Myelogenous Leukemia Cells via JNK-Mediated p62/SQSTM1 Expression and AMPK Activation
- Author
-
Patrick Auberger, Sophie Raynaud, Jill-Patrice Cassuto, Frederic Luciano, Nina Fenouille, Guillaume Robert, and Alexandre Puissant
- Abstract
Supplementary Figure 3 from Resveratrol Promotes Autophagic Cell Death in Chronic Myelogenous Leukemia Cells via JNK-Mediated p62/SQSTM1 Expression and AMPK Activation
- Published
- 2023
- Full Text
- View/download PDF
29. MCB-613 exploits a collateral sensitivity in drug resistantEGFR-mutant non-small cell lung cancer through covalent inhibition of KEAP1
- Author
-
Christopher F. Bassil, Gray R. Anderson, Benjamin Mayro, Kayleigh N. Askin, Peter S. Winter, Samuel Gruber, Tierney M. Hall, Jacob P. Hoj, Christian Cerda-Smith, Haley M. Hutchinson, Shane T. Killarney, Katherine R. Singleton, Li Qin, Kévin Jubien-Girard, Cécile Favreau, Anthony R. Martin, Guillaume Robert, Rachid Benhida, Patrick Auberger, Ann Marie Pendergast, David M. Lonard, Alexandre Puissant, and Kris C. Wood
- Abstract
Targeted therapies have revolutionized cancer chemotherapy. Unfortunately, most patients develop multifocal resistance to these drugs within a matter of months. Here, we used a high-throughput phenotypic small molecule screen to identify MCB-613 as a compound that selectively targetsEGFR-mutant, EGFR inhibitor-resistant non-small cell lung cancer (NSCLC) cells harboring diverse resistance mechanisms. Subsequent proteomic and functional genomic screens involving MCB-613 identified its target in this context to be KEAP1, revealing that this gene is selectively essential in the setting of EGFR inhibitor resistance. In-depth molecular characterization demonstrated that (1) MCB-613 binds KEAP1 covalently; (2) a single molecule of MCB-613 is capable of bridging two KEAP1 monomers together; and, (3) this modification interferes with the degradation of canonical KEAP1 substrates such as NRF2. Surprisingly, NRF2 knockout sensitizes cells to MCB-613, suggesting that the drug functions through modulation of an alternative KEAP1 substrate. Together, these findings advance MCB-613 as a new tool for exploiting the selective essentiality of KEAP1 in drug-resistant,EGFR-mutant NSCLC cells.
- Published
- 2023
- Full Text
- View/download PDF
30. Walking the Tightrope: Balancing Delicate Inflammation Response to Eradicate Acute Myeloid Leukemia
- Author
-
Alexandre Puissant and Hind Medyouf
- Subjects
Inflammation ,Leukemia, Myeloid, Acute ,Oncology ,Humans ,Monocytes ,Article - Abstract
Leukemic blasts are immune cells gone awry. We hypothesized that dysregulation of inflammatory pathways contributes to the maintenance of their leukemic state and can be exploited as a cell-intrinsic, self-directed immunotherapy. To this end, we applied genome-wide screens to discover genetic vulnerabilities in acute myeloid leukemia (AML) cells implicated in inflammatory pathways. We identified the immune modulator interferon regulatory factor 2 binding protein 2 (IRF2BP2) as a selective AML dependency. We validated AML cell dependency on IRF2BP2 with genetic and protein degradation approaches in vitro and genetically in vivo. Chromatin and global gene expression studies demonstrated that IRF2BP2 represses IL-1ß/TNFα signaling via NF-κB, and IRF2BP2 perturbation results in an acute inflammatory state leading to AML cell death. These findings elucidate a hitherto unexplored AML dependency, reveal cell-intrinsic inflammatory signaling as a mechanism priming leukemic blasts for regulated cell death, and establish IRF2BP2-mediated transcriptional repression as a mechanism for blast survival.
- Published
- 2022
31. Tumor Lysis Syndrome and AKI: Beyond Crystal Mechanisms
- Author
-
Marine Arnaud, Maud Loiselle, Camille Vaganay, Stéphanie Pons, Emmanuel Letavernier, Jordane Demonchy, Sofiane Fodil, Manal Nouacer, Sandrine Placier, Perrine Frère, Eden Arrii, Julien Lion, Nuala Mooney, Raphael Itzykson, Chakib Djediat, Alexandre Puissant, Lara Zafrani, and INSERM, CEA, U976 - HIPI, Institut de Recherche Saint Louis, Université de Paris, Paris, France
- Subjects
Basic Research ,Nephrology ,[SDV]Life Sciences [q-bio] ,Humans ,General Medicine ,Acute Kidney Injury ,Tumor Lysis Syndrome ,Uric Acid - Abstract
International audience; Background The pathophysiology of AKI during tumor lysis syndrome (TLS) is not well understood due to the paucity of data. We aimed to decipher crystal-dependent and crystal-independent mechanisms of TLS-induced AKI. Methods Crystalluria, plasma cytokine levels, and extracellular histones levels were measured in two cohorts of patients with TLS. We developed a model of TLS in syngeneic mice with acute myeloid leukemia, and analyzed ultrastructural changes in kidneys and endothelial permeability using intravital confocal microscopy. In parallel, we studied the endothelial toxicity of extracellular histones in vitro. Results The study provides the first evidence that previously described crystal-dependent mechanisms are insufficient to explain TLS-induced AKI. Extracellular histones that are released in huge amounts during TLS caused profound endothelial alterations in the mouse model. The mechanisms of histone-mediated damage implicates endothelial cell activation mediated by Toll-like receptor 4. Heparin inhibits extracellular histones and mitigates endothelial dysfunction during TLS. Conclusion This study sheds new light on the pathophysiology of TLS-induced AKI and suggests that extracellular histones may constitute a novel target for therapeutic intervention in TLS when endothelial dysfunction occurs.
- Published
- 2022
- Full Text
- View/download PDF
32. Using antagonistic pleiotropy to design a chemotherapy-induced evolutionary trap to target drug resistance in cancer
- Author
-
Abigail Xie, Yeong-ran Ahn, Lorin Crawford, Peter S. Winter, Bryann Pardieu, Justine C. Rutter, Kevin H. Lin, Raiyan T. Sobhan, Antoine Forget, Katherine R. Singleton, Jason W. Locasale, Alexandre Puissant, Grace R. Anderson, Raphael Itzykson, Kris C. Wood, Amy E. Decker, Ziwei Dai, Reinaldo Dal Bello, and Emily T. Winn
- Subjects
Quantitative Trait Loci ,HL-60 Cells ,Drug resistance ,Environment ,Biology ,Article ,Cell Line ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Pleiotropy ,Cell Line, Tumor ,Neoplasms ,Genetics ,Animals ,Humans ,030304 developmental biology ,Maladaptation ,0303 health sciences ,Nuclear Proteins ,Myeloid leukemia ,Genetic Pleiotropy ,Adaptation, Physiological ,Biological Evolution ,Phenotype ,3. Good health ,Bromodomain ,HEK293 Cells ,Drug Resistance, Neoplasm ,Evolutionary trap ,Genetic Fitness ,CRISPR-Cas Systems ,Adaptation ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
Local adaptation directs populations towards environment-specific fitness maxima through acquisition of positively selected traits. However, rapid environmental changes can identify hidden fitness trade-offs that turn adaptation into maladaptation, resulting in evolutionary traps. Cancer, a disease that is prone to drug resistance, is in principle susceptible to such traps. We therefore performed pooled CRISPR-Cas9 knockout screens in acute myeloid leukemia (AML) cells treated with various chemotherapies to map the drug-dependent genetic basis of fitness trade-offs, a concept known as antagonistic pleiotropy (AP). We identified a PRC2-NSD2/3-mediated MYC regulatory axis as a drug-induced AP pathway whose ability to confer resistance to bromodomain inhibition and sensitivity to BCL-2 inhibition templates an evolutionary trap. Across diverse AML cell-line and patient-derived xenograft models, we find that acquisition of resistance to bromodomain inhibition through this pathway exposes coincident hypersensitivity to BCL-2 inhibition. Thus, drug-induced AP can be leveraged to design evolutionary traps that selectively target drug resistance in cancer.
- Published
- 2020
- Full Text
- View/download PDF
33. Cystine Uptake Inhibition Potentiates Front-Line Therapies In Acute Myeloid Leukemia
- Author
-
Bryann Pardieu, Justine Pasanisi, Frank Ling, Reinaldo Dal Bello, Justine Penneroux, Angela Su, Romane Joudinaud, Laureen Chat, Hsin Chieh Wu, Matthieu Duchmann, Gaetano Sodaro, Clémentine Chauvel, Florence A. Castelli, Loic Vasseur, Kim Pacchiardi, Yannis Belloucif, Marie-Charlotte Laiguillon, Eshwar Meduri, Camille Vaganay, Gabriela Alexe, Jeannig Berrou, Chaima Benaksas, Antoine Forget, Thorsten Braun, Claude Gardin, Emmanuel Raffoux, Emmanuelle Clappier, Lionel Adès, Hugues de Thé, François Fenaille, Brian J. Huntly, Kimberly Stegmaier, Hervé Dombret, Nina Fenouille, Camille Lobry, Alexandre Puissant, Raphael Itzykson, Génomes, biologie cellulaire et thérapeutiques (GenCellDi (U944 / UMR7212)), Collège de France (CdF (institution))-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), Centre interdisciplinaire de recherche en biologie (CIRB), Labex MemoLife, École normale supérieure - Paris (ENS-PSL), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Collège de France (CdF (institution))-Ecole Superieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Université Paris sciences et lettres (PSL)-École normale supérieure - Paris (ENS-PSL), Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Hôpital Henri Mondor, Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay-Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), MetaboHUB, Pathologies articulaires associées aux maladies métaboliques et à l’âge [CRSA], Centre de Recherche Saint-Antoine (CRSA), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), Harvard Medical School [Boston] (HMS), Institut de Recherche Saint-Louis - Hématologie Immunologie Oncologie (Département de recherche de l’UFR de médecine, ex- Institut Universitaire Hématologie-IUH) (IRSL), Université Paris Cité (UPCité), Signalisation, radiobiologie et cancer, Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS), Hôpital Avicenne [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Hopital Saint-Louis [AP-HP] (AP-HP), Hôpitaux Universitaires Saint-Louis, Lariboisière, Fernand-Widal, Collège de France - Chaire Oncologie cellulaire et moléculaire, Collège de France (CdF (institution))-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité)-Collège de France (CdF (institution))-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), Belloucif, Yannis [0000-0001-6364-4098], Alexe, Gabriela [0000-0002-5668-6297], Adès, Lionel [0000-0002-9020-8766], de Thé, Hugues [0000-0002-1113-4472], Fenaille, François [0000-0001-6787-4149], Huntly, Brian J [0000-0003-0312-161X], Stegmaier, Kimberly [0000-0003-0218-7895], Lobry, Camille [0000-0003-0550-4921], Puissant, Alexandre [0000-0002-3997-9282], Itzykson, Raphael [0000-0003-2139-6262], and Apollo - University of Cambridge Repository
- Subjects
Acute Myeloid Leukemia ,Cancer Research ,Acute Myeloid Leukemia Cysteine Ferroptosis Drug Repurposing ,[SDV]Life Sciences [q-bio] ,Daunorubicin ,Nuclear Proteins ,Hematology ,Sulfasalazine ,Leukemia, Myeloid, Acute ,Drug Repurposing ,Oncology ,hemic and lymphatic diseases ,Cell Line, Tumor ,Cystine ,Humans ,Ferroptosis ,Cysteine ,neoplasms - Abstract
By querying metabolic pathways associated with leukemic stemness and survival in multiple AML datasets, we nominated SLC7A11 encoding the xCT cystine importer as a putative AML dependency. Genetic and chemical inhibition of SLC7A11 impaired the viability and clonogenic capacity of AML cell lines in a cysteine-dependent manner. Sulfasalazine, a broadly available drug with xCT inhibitory activity, had anti-leukemic activity against primary AML samples in ex vivo cultures. Multiple metabolic pathways were impacted upon xCT inhibition, resulting in depletion of glutathione pools in leukemic cells and oxidative stress-dependent cell death, only in part through ferroptosis. Higher expression of cysteine metabolism genes and greater cystine dependency was noted in NPM1-mutated AMLs. Among eight anti-leukemic drugs, the anthracycline daunorubicin was identified as the top synergistic agent in combination with sulfasalazine in vitro. Addition of sulfasalazine at a clinically relevant concentration significantly augmented the anti-leukemic activity of a daunorubicin-cytarabine combination in a panel of 45 primary samples enriched in NPM1-mutated AML. These results were confirmed in vivo in a patient-derived xenograft model. Collectively, our results nominate cystine import as a druggable target in AML and raise the possibility to repurpose sulfasalazine for the treatment of AML, notably in combination with chemotherapy.
- Published
- 2022
- Full Text
- View/download PDF
34. JAK Inhibition Mediates Clonal Selection of RAS Pathway Mutations in Myeloproliferative Neoplasms
- Author
-
Nabih Maslah, Blandine Roux, Nina Kaci, Emmanuelle Verger, Rafael Daltro De Oliveira, Hélène Pasquer, Nicolas Gauthier, Juliette Soret, Saravanan Ganesan, Panhong Gou, Frank Ling, Nathalie Parquet, William Vainchenker, Emmanuel Raffoux, Rose Ann Padua, Stephane Giraudier, Alexandre Puissant, Camille Lobry, Jean-Jacques Kiladjian, Bruno Cassinat, and Lina Benajiba
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
35. Screening of ETO2-GLIS2-induced Super Enhancers identifies targetable cooperative dependencies in acute megakaryoblastic leukemia
- Author
-
Salima Benbarche, Cécile K. Lopez, Eralda Salataj, Zakia Aid, Cécile Thirant, Marie-Charlotte Laiguillon, Séverine Lecourt, Yannis Belloucif, Camille Vaganay, Marion Antonini, Jiang Hu, Alexandra da Silva Babinet, Delphine Ndiaye-Lobry, Bryann Pardieu, Arnaud Petit, Alexandre Puissant, Julie Chaumeil, Thomas Mercher, and Camille Lobry
- Subjects
Multidisciplinary - Abstract
Super Enhancers (SEs) are clusters of regulatory elements associated with cell identity and disease. However, whether these elements are induced by oncogenes and can regulate gene modules cooperating for cancer cell transformation or maintenance remains elusive. To address this question, we conducted a genome-wide CRISPRi-based screening of SEs in ETO2-GLIS2+acute megakaryoblastic leukemia. This approach revealed SEs essential for leukemic cell growth and survival that are induced by ETO2-GLIS2 expression. In particular, we identified a de novo SE specific of this leukemia subtype and regulating expression of tyrosine kinase–associated receptorsKITandPDGFRA. Combined expression of these two receptors was required for leukemic cell growth, and CRISPRi-mediated inhibition of this SE or treatment with tyrosine kinase inhibitors impaired progression of leukemia in vivo in patient-derived xenografts experiments. Our results show that fusion oncogenes, such as ETO2-GLIS2, can induce activation of SEs regulating essential gene modules synergizing for leukemia progression.
- Published
- 2022
36. A multiparametric niche-like drug screening platform in acute myeloid leukemia
- Author
-
Reinaldo Dal Bello, Justine Pasanisi, Romane Joudinaud, Matthieu Duchmann, Bryann Pardieu, Paolo Ayaka, Giuseppe Di Feo, Gaetano Sodaro, Clémentine Chauvel, Rathana Kim, Loic Vasseur, Laureen Chat, Frank Ling, Kim Pacchiardi, Camille Vaganay, Jeannig Berrou, Chaima Benaksas, Nicolas Boissel, Thorsten Braun, Claude Preudhomme, Hervé Dombret, Emmanuel Raffoux, Nina Fenouille, Emmanuelle Clappier, Lionel Adès, Alexandre Puissant, and Raphael Itzykson
- Subjects
Leukemia, Myeloid, Acute ,Oncology ,Cytarabine ,Drug Evaluation, Preclinical ,Neoplastic Stem Cells ,Humans ,Anthracyclines ,Mesenchymal Stem Cells ,Hematology - Abstract
Functional precision medicine in AML often relies on short-term in vitro drug sensitivity screening (DSS) of primary patient cells in standard culture conditions. We designed a niche-like DSS assay combining physiologic hypoxia (O2 3%) and mesenchymal stromal cell (MSC) co-culture with multiparameter flow cytometry to enumerate lymphocytes and differentiating (CD11/CD14/CD15+) or leukemic stem cell (LSC)-enriched (GPR56+) cells within the leukemic bulk. After functional validation of GPR56 expression as a surrogate for LSC enrichment, the assay identified three patterns of response, including cytotoxicity on blasts sparing LSCs, induction of differentiation, and selective impairment of LSCs. We refined our niche-like culture by including plasma-like amino-acid and cytokine concentrations identified by targeted metabolomics and proteomics of primary AML bone marrow plasma samples. Systematic interrogation revealed distinct contributions of each niche-like component to leukemic outgrowth and drug response. Short-term niche-like culture preserved clonal architecture and transcriptional states of primary leukemic cells. In a cohort of 45 AML samples enriched for NPM1c AML, the niche-like multiparametric assay could predict morphologically (p = 0.02) and molecular (NPM1c MRD, p = 0.04) response to anthracycline-cytarabine induction chemotherapy. In this cohort, a 23-drug screen nominated ruxolitinib as a sensitizer to anthracycline-cytarabine. This finding was validated in an NPM1c PDX model.
- Published
- 2021
37. Endothelial cells: major players in acute myeloid leukaemia
- Author
-
Sofiane Fodil, Marine Arnaud, Camille Vaganay, Alexandre Puissant, Etienne Lengline, Nuala Mooney, Raphael Itzykson, Lara Zafrani, and INSERM, CEA, U976 - HIPI, Institut de Recherche Saint Louis, Université de Paris, Paris, France
- Subjects
Clonal Evolution ,Leukemia, Myeloid, Acute ,Oncology ,Bone Marrow ,[SDV]Life Sciences [q-bio] ,Tumor Microenvironment ,Endothelial Cells ,Humans ,Bone Marrow Cells ,Hematology - Abstract
The role of the vascular microenvironment is increasingly studied in acute myeloid leukaemia (AML). Complex interactions between endothelial cells (ECs) and pre-leukaemic cells may contribute to the clonal evolution of pre-leukaemic stem cells in the bone marrow niche and to the proliferation, survival and chemoresistance of leukaemic cells. Through the expression of different adhesion molecules, ECs play a key role in the development of specific acute complications of AML, including leukostasis, acute respiratory failure, acute kidney injury or neurological complications. Moreover, in newly diagnosed patients, leukaemic cells promote endothelial activation and subsequent disseminated intravascular coagulation. Mechanisms of this bi-directional dialogue between leukaemic cells and ECs will reveal possible therapeutic targets to be explored to improve the survival of AML patients.
- Published
- 2021
- Full Text
- View/download PDF
38. Matched Targeted Therapy for Pediatric Patients with Relapsed, Refractory, or High-Risk Leukemias: A Report from the LEAP Consortium
- Author
-
Cristina E. Tognon, Alma Imamovic, Peter D. Cole, Anjali Cremer, Todd M. Cooper, Alexandre Puissant, Catherine Clinton, Asmani A. Adhav, Patrick A. Brown, Kristen E. Stevenson, Mignon L. Loh, Justine M. Kahn, Nathan Gossai, Elliot Stieglitz, Wilian A. Cortopassi, Andrew E. Place, Stephen P. Hunger, Michael J. Burke, Lewis B. Silverman, Annette S. Kim, Nicole Ocasio-Martinez, Diego Garrido Ruiz, Jeffrey W. Tyner, Matthew J. Barth, Lisa M. Gennarini, Yana Pikman, Neal I. Lindeman, Maria Luisa Sulis, Lia Gore, Beth Apsel Winger, Neekesh V. Dharia, Traci M. Blonquist, Yuting Li, Kimberly Stegmaier, Marian H. Harris, Jeffrey A. Magee, Katherine Tarlock, Neerav Shukla, Melinda Pauly, Kelly W. Maloney, Matthew P. Jacobson, Angela Su, Tasleema Patel, Giacomo Gotti, Cristina F. Contreras, Shan Lin, Haley L. Faust, Amanda L. Robichaud, Jing Chen, Sarah K. Tasian, Katherine A. Janeway, Amy Saur Conway, and Jennifer L. McNeer
- Subjects
0301 basic medicine ,Oncology ,Male ,medicine.medical_treatment ,Targeted therapy ,Cohort Studies ,0302 clinical medicine ,2.1 Biological and endogenous factors ,Prospective Studies ,Molecular Targeted Therapy ,Aetiology ,Child ,Cancer ,Pediatric ,Leukemia ,Tumor ,Hematology ,Local ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Disease Progression ,Female ,Development of treatments and therapeutic interventions ,Biotechnology ,medicine.medical_specialty ,Pediatric Cancer ,Childhood Leukemia ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,MEDLINE ,03 medical and health sciences ,Rare Diseases ,Refractory ,Clinical Research ,Internal medicine ,Biomarkers, Tumor ,medicine ,Genetics ,Humans ,business.industry ,Human Genome ,medicine.disease ,Precision medicine ,United States ,Clinical trial ,030104 developmental biology ,Neoplasm Recurrence ,Orphan Drug ,Good Health and Well Being ,Relapsed refractory ,Feasibility Studies ,Neoplasm Recurrence, Local ,business ,Biomarkers - Abstract
Despite a remarkable increase in the genomic profiling of cancer, integration of genomic discoveries into clinical care has lagged behind. We report the feasibility of rapid identification of targetable mutations in 153 pediatric patients with relapsed/refractory or high-risk leukemias enrolled on a prospective clinical trial conducted by the LEAP Consortium. Eighteen percent of patients had a high confidence Tier 1 or 2 recommendation. We describe clinical responses in the 14% of patients with relapsed/refractory leukemia who received the matched targeted therapy. Further, in order to inform future targeted therapy for patients, we validated variants of uncertain significance, performed ex vivo drug-sensitivity testing in patient leukemia samples, and identified new combinations of targeted therapies in cell lines and patient-derived xenograft models. These data and our collaborative approach should inform the design of future precision medicine trials. Significance: Patients with relapsed/refractory leukemias face limited treatment options. Systematic integration of precision medicine efforts can inform therapy. We report the feasibility of identifying targetable mutations in children with leukemia and describe correlative biology studies validating therapeutic hypotheses and novel mutations. See related commentary by Bornhauser and Bourquin, p. 1322. This article is highlighted in the In This Issue feature, p. 1307
- Published
- 2021
39. Targeting acute myeloid leukemia dependency on VCP-mediated DNA repair through a selective second-generation small-molecule inhibitor
- Author
-
Amy Saur Conway, Lina Benajiba, Camille Vaganay, Daniel J. DeAngelo, Olivier Hermine, Yangzhong Tang, Kasper Lage, Han-Jie Zhou, Ilene Galinsky, Patrick Auberger, Jesse D. Vargas, Richard Stone, Josée Guirouilh-Barbat, Gabriela Alexe, Chaïma Benaksas, Yana Pikman, Alexandre Puissant, Bryann Pardieu, Daniel Anderson, Kimberly Stegmaier, Blandine Roux, Jana M. Ellegast, Bernard S. Lopez, Monica Schenone, Christina R. Hartigan, Michael T. Hemann, Linda Ross, Nina Fenouille, Mehdi Khaled, Frank Ling, Edyta Malolepsza, Frederic Luciano, Steven A. Carr, Gaetano Sodaro, Ronan Le Moigne, Tony Wu, [Institut Cochin] Département Développement, Reproduction et Cancer (DRC), Institut Cochin (IC UM3 (UMR 8104 / U1016)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), Génomes, biologie cellulaire et thérapeutiques (GenCellDi (UMR_S_944)), Collège de France (CdF (institution))-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), and Harvard Medical School [Boston] (HMS)
- Subjects
Myeloid ,DNA Repair ,DNA repair ,Valosin-containing protein ,[SDV]Life Sciences [q-bio] ,Antineoplastic Agents ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Article ,Small hairpin RNA ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Valosin Containing Protein ,hemic and lymphatic diseases ,Cell Line, Tumor ,medicine ,Animals ,Humans ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,Adenosine Triphosphatases ,0303 health sciences ,biology ,Cancer ,Myeloid leukemia ,General Medicine ,medicine.disease ,3. Good health ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,Cancer research - Abstract
The development and survival of cancer cells require adaptive mechanisms to stress. Such adaptations can confer intrinsic vulnerabilities, enabling the selective targeting of cancer cells. Through a pooled in vivo short hairpin RNA (shRNA) screen, we identified the adenosine triphosphatase associated with diverse cellular activities (AAA-ATPase) valosin-containing protein (VCP) as a top stress-related vulnerability in acute myeloid leukemia (AML). We established that AML was the most responsive disease to chemical inhibition of VCP across a panel of 16 cancer types. The sensitivity to VCP inhibition of human AML cell lines, primary patient samples, and syngeneic and xenograft mouse models of AML was validated using VCP-directed shRNAs, overexpression of a dominant-negative VCP mutant, and chemical inhibition. By combining mass spectrometry-based analysis of the VCP interactome and phospho-signaling studies, we determined that VCP is important for ataxia telangiectasia mutated (ATM) kinase activation and subsequent DNA repair through homologous recombination in AML. A second-generation VCP inhibitor, CB-5339, was then developed and characterized. Efficacy and safety of CB-5339 were validated in multiple AML models, including syngeneic and patient-derived xenograft murine models. We further demonstrated that combining DNA-damaging agents, such as anthracyclines, with CB-5339 treatment synergizes to impair leukemic growth in an MLL-AF9-driven AML murine model. These studies support the clinical testing of CB-5339 as a single agent or in combination with standard-of-care DNA-damaging chemotherapy for the treatment of AML.
- Published
- 2021
- Full Text
- View/download PDF
40. Drug Resistance in Hematological Malignancies
- Author
-
Patrick Auberger, Jerome Tamburini-Bonnefoy, and Alexandre Puissant
- Subjects
Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,MEDLINE ,Drug resistance ,Immunotherapy, Adoptive ,Catalysis ,Inorganic Chemistry ,lcsh:Chemistry ,Pharmacotherapy ,Text mining ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Combined Modality Therapy ,Humans ,Molecular Targeted Therapy ,Physical and Theoretical Chemistry ,Molecular Biology ,lcsh:QH301-705.5 ,Spectroscopy ,business.industry ,Organic Chemistry ,General Medicine ,Immunotherapy ,Computer Science Applications ,Editorial ,medicine.anatomical_structure ,n/a ,lcsh:Biology (General) ,lcsh:QD1-999 ,Drug Resistance, Neoplasm ,Hematologic Neoplasms ,Drug Therapy, Combination ,Bone marrow ,business - Abstract
Hematological malignancies define a highly heterogeneous set of blood-, bone marrow-, and organ-associated diseases with highly variable prognoses that constantly relapse upon treatment [...]
- Published
- 2020
41. Characterization of midostaurin as a dual inhibitor of FLT3 and SYK and potentiation of FLT3 inhibition against FLT3-ITD-driven leukemia harboring activated SYK kinase
- Author
-
Kimberly Stegmaier, David M. Weinstock, Richard Stone, Amanda L. Christie, Alexandre Puissant, Ellen Weisberg, Paul W. Manley, Renee Wright, Jing Yang, Martin Sattler, Chengcheng Meng, John F. Daley, Sara J. Buhrlage, Suzan Lazo, James D. Griffin, and Michael J Buonopane
- Subjects
0301 basic medicine ,FLT3-ITD ,acute myelogenous leukemia ,Syk ,R406 ,03 medical and health sciences ,chemistry.chemical_compound ,fluids and secretions ,0302 clinical medicine ,hemic and lymphatic diseases ,R788 ,medicine ,Midostaurin ,Quizartinib ,business.industry ,Myeloid leukemia ,hemic and immune systems ,medicine.disease ,Leukemia ,midostaurin ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,embryonic structures ,Immunology ,Cancer research ,biological phenomena, cell phenomena, and immunity ,business ,FLT3 Inhibitor ,Tyrosine kinase ,Priority Research Paper ,Crenolanib - Abstract
Oncogenic FLT3 kinase is a clinically validated target in acute myeloid leukemia (AML), and both multi-targeted and selective FLT3 inhibitors have been developed. Spleen tyrosine kinase (SYK) has been shown to be activated and increased in FLT3-ITD-positive AML patients, and has further been shown to be critical for transformation and maintenance of the leukemic clone in these patients. Further, over-expression of constitutively activated SYK causes resistance to highly selective FLT3 tyrosine kinase inhibitors (TKI). Up to now, the activity of the multi-targeted FLT3 inhibitor, midostaurin, against cells expressing activated SYK has not been explored in the context of leukemia, although SYK has been identified as a target of midostaurin in systemic mastocytosis. We compared the ability of midostaurin to inhibit activated SYK in mutant FLT3-positive AML cells with that of inhibitors displaying dual SYK/FLT3 inhibition, targeted SYK inhibition, and targeted FLT3 inhibition. Our findings suggest that dual FLT3/SYK inhibitors and FLT3-targeted drugs potently kill oncogenic FLT3-transformed cells, while SYK-targeted small molecule inhibition displays minimal activity. However, midostaurin and other dual FLT3/SYK inhibitors display superior anti-proliferative activity when compared to targeted FLT3 inhibitors, such as crenolanib and quizartinib, against cells co-expressing FLT3-ITD and constitutively activated SYK-TEL. Interestingly, additional SYK suppression potentiated the effects of dual FLT3/SYK inhibitors and targeted FLT3 inhibitors against FLT3-ITD-driven leukemia, both in the absence and presence of activated SYK. Taken together, our findings have important implications for the design of drug combination studies in mutant FLT3-positive patients and for the design of future generations of FLT3 inhibitors.
- Published
- 2017
- Full Text
- View/download PDF
42. L’autophagie, acteur clé de la leucémogenèse et cible thérapeutique dans les hémopathies malignes
- Author
-
Guillaume Robert, Alexandre Puissant, Frederic Luciano, Patrick Auberger, and Arnaud Jacquel
- Subjects
0301 basic medicine ,03 medical and health sciences ,030104 developmental biology ,General Medicine ,Biology ,General Biochemistry, Genetics and Molecular Biology - Published
- 2017
- Full Text
- View/download PDF
43. Niche-like Ex Vivo High Throughput (NEXT) Drug Screening Platform in Acute Myeloid Leukemia
- Author
-
Loic Vasseur, Lionel Ades, Camille Vaganay, Lionel Faivre, Matthieu Duchmann, Kim Pacchiardi, Clémentine Chauvel, Raphael Itzykson, Bryann Pardieu, Chaïma Benaksas, Alexandre Puissant, Reinaldo Dal Bello Figueiras, Gaetano Sodaro, Frank Ling, Emmanuel Raffoux, Justine Pasanisi, Claude Preudhomme, Romane Joudinaud, and Emmanuelle Clappier
- Subjects
Oncology ,medicine.medical_specialty ,Ruxolitinib ,NPM1 ,business.industry ,Venetoclax ,Immunology ,Context (language use) ,Cell Biology ,Hematology ,Precision medicine ,Biochemistry ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,In vivo ,Internal medicine ,Medicine ,Bone marrow ,business ,health care economics and organizations ,Ex vivo ,medicine.drug - Abstract
Context. Functional precision medicine is gaining momentum in AML, notably through ex vivo drug sensitivity screening (DSS) of primary patient (pt) cells (Pemovska Cancer Discov 2013, Tyner Nature 2018). The DSS landscape differs across genetic AML subgroups (Tyner Nature 2016), of which NPM1mut is the most frequent (Papaemmanuil NEJM 2016). DSS in AML has mostly been done in standard conditions, with overall viability as unique endpoint. Niche signals, which can be partly mimicked in vitro, convey drug resistance in vivo. Drugs can induce a variety of cell fates in AML. Induction of differentiation rather than killing of blasts, can result in false negative results in global viability assays. Persistence of leukemic stem cells (LSC) represents a major cause of treatment failure. GPR56 is a ubiquitous surface marker enriching for LSCs and stable upon short-term ex vivo culture (Pabst Blood 2016). Objectives. To develop an ex vivo niche-like multiparametric DSS platform for primary AML cells. To validate its clinical relevance in NPM1mut pts treated with conventional DNR-AraC chemotherapy. To discover new sensitizers to DNR-AraC chemotherapy in NPM1mut AML. Results. We designed an MFC panel to count viable blasts and measure their differentiation (CD11b/CD14/CD15) and stemness (GPR56) after exclusion of residual lymphocytes (Figure 1A). We validated GPR56 expression as stemness marker based on increased retention of GPR56+ cells in niche-like coculture combining hypoxia (O2 3%) and MSC compared to standard conditions (p To further mimic in vivo conditions, we derived a MEMa-based plasma-like medium (PLM) based on targeted metabolomics (Figure 1E) and electro-chemoluminescent cytokine assays of 29 diagnostic AML bone marrow plasma samples compared to conditioned media of primary AML cells cultured in niche-like conditions (MSC, hypoxia). This instructed the design of our custom PLM with dialyzed FBS and defined low-dose (~1 ng/mL range) cytokines (CK) and amino-acid (AA) concentrations. We next investigated the contribution of MSCs, hypoxia, plasma-like AAs and CKs on blasts viability, differentiation, stemness and drug response in 3 NPM1mut AMLs exposed to fixed concentrations of 6 core AML therapies. This analysis uncovered significant interactions between these 4 niche components in dictating blast viability and stemness upon 72h ex vivo culture (Figure 1F) and revealed the distinct contribution of these niche components to drug sensitivity. RNA-seq of primary blasts cultured in niche-like, plasma-like conditions revealed marked enrichment of stemness pathways compared to ex vivo culture in standard conditions. Finally, we explored DNR-AraC (five-point serial dilution) alone or in combination with fixed, clinically relevant concentrations of 24 drugs in 49 primary AML samples (including 34 NPM1mut). Using AUCs of DNR-AraC on lymphocytes as internal control, we first validated our NEXT assay on NPM1 MRD levels in the 34 NPM1mut pts treated frontline with conventional DNR-AraC regimens (Figure 1G). Across all 49 pts, we uncovered 11 different optimal 'third-drugs', stressing the role of our NEXT assay to deploy precision medicine in daily practice. At the population level, we could nominate 3 top combinations, two of which are currently in clinical investigation (Venetoclax and Selinexor). The unpublished sensitizing effect of low dose (0.25µM) Ruxolitinib on DNR-AraC uncovered with our NEXT assay is currently being investigated in PDX models. Conclusion. We designed the NEXT assay, a multiparametric drug screening of AML viability, differentiation and stemness in niche-like culture combining hypoxia, stromal interactions and plasma-like medium. Components of the niche-like culture interact to govern leukemic viability and stemness. Our assay could predict MRD achievement in NPM1mut AML and identifies novel sensitizers to DNR-AraC in these pts. Disclosures Clappier: Amgen: Honoraria, Research Funding. Ades:Abbvie: Honoraria, Membership on an entity's Board of Directors or advisory committees; takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees; jazz: Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Research Funding; novartis: Research Funding; Celgene/BMS: Research Funding. Itzykson:Amgen: Membership on an entity's Board of Directors or advisory committees; Otsuka Pharma: Membership on an entity's Board of Directors or advisory committees; Jazz Pharmaceuticals: Honoraria, Membership on an entity's Board of Directors or advisory committees; Stemline: Membership on an entity's Board of Directors or advisory committees; Oncoethix (now Merck): Research Funding; Janssen: Research Funding; Karyopharm: Membership on an entity's Board of Directors or advisory committees; Abbvie: Honoraria; Daiichi Sankyo: Honoraria; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; BMS (Celgene): Honoraria; Sanofi: Honoraria; Astellas: Honoraria.
- Published
- 2020
- Full Text
- View/download PDF
44. Targeting serine hydroxymethyltransferases 1 and 2 for T-cell acute lymphoblastic leukemia therapy
- Author
-
Amy Saur Conway, Jun Qi, Giovanni Roti, Amanda L. Robichaud, Yana Pikman, Alexandre Puissant, Caroline A. Lewis, Gabriela Alexe, Nicole Ocasio-Martinez, Kimberly Stegmaier, Samuel Kitara, Matthew G. Vander Heiden, Angela Su, and Frances F. Diehl
- Subjects
chemistry.chemical_classification ,Purine ,0303 health sciences ,Cell cycle checkpoint ,business.industry ,T cell ,medicine.disease ,3. Good health ,03 medical and health sciences ,Metabolic pathway ,Leukemia ,chemistry.chemical_compound ,0302 clinical medicine ,Enzyme ,medicine.anatomical_structure ,chemistry ,In vivo ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,business ,030304 developmental biology - Abstract
Despite progress in the treatment of acute lymphoblastic leukemia (ALL), T-cell ALL (T-ALL) has limited treatment options particularly in the setting of relapsed/refractory disease. Using an unbiased genome-scale CRISPR-Cas9 screen we sought to identify pathway dependencies for T-ALL which could be harnessed for therapy development. Disruption of the one-carbon folate, purine and pyrimidine pathways scored as the top metabolic pathways required for T-ALL proliferation. We used a recently developed inhibitor of SHMT1 and SHMT2, RZ-2994, to characterize the effect of inhibiting these enzymes of the one-carbon folate pathway in T-ALL and found that T-ALL cell lines were differentially sensitive to RZ-2994, with a S/G2 cell cycle arrest. The effects of SHMT1/2 inhibition were rescued by formate supplementation. Loss of both SHMT1 and SHMT2 was necessary for impaired growth and cell cycle arrest, with suppression of both SHMT1 and SHMT2 impairing leukemia progressionin vivo. RZ-2994 decreased leukemia burdenin vivoand remained effective in the setting of methotrexate resistancein vitro. This study highlights the significance of the one-carbon folate pathway in T-ALL and supports further development of SHMT inhibitors for treatment of T-ALL and other cancers.
- Published
- 2020
- Full Text
- View/download PDF
45. CDK6 is an essential direct target of NUP98 fusion proteins in acute myeloid leukemia
- Author
-
Florian Grebien, Ha Thi Thanh Pham, Arnaud Petit, Gabriele Manhart, Roland Meisel, Alexandre Puissant, Peter Valent, Johannes Schmoellerl, Luisa Schmidt, Selina Troester, Mohanty Sagarajit, Hélène Lapillonne, Raphael Itzykson, Inês Amorim Monteiro Barbosa, Gregor Hoermann, Michael Heuser, Jessica Ebner, Nicolas Duployez, Tania Brandstoetter, Johannes Zuber, Ezgi Aslan, Thomas Eder, Richard Moriggl, Stefan Terlecki-Zaniewicz, Christa Van Der Veen, Veronika Sexl, and Barbara Maurer
- Subjects
Myeloid ,Oncogene Proteins ,Oncogene Proteins, Fusion ,Immunology ,Biochemistry ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Drug Delivery Systems ,medicine ,Animals ,Humans ,030304 developmental biology ,0303 health sciences ,biology ,Gene Expression Profiling ,Myeloid leukemia ,Cell Biology ,Hematology ,Cyclin-Dependent Kinase 6 ,medicine.disease ,Fusion protein ,3. Good health ,Chromatin ,Gene expression profiling ,Nuclear Pore Complex Proteins ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Cyclin-dependent kinase 6 - Abstract
Fusion proteins involving Nucleoporin 98 (NUP98) are recurrently found in acute myeloid leukemia (AML) and are associated with poor prognosis. Lack of mechanistic insight into NUP98-fusion–dependent oncogenic transformation has so far precluded the development of rational targeted therapies. We reasoned that different NUP98-fusion proteins deregulate a common set of transcriptional targets that might be exploitable for therapy. To decipher transcriptional programs controlled by diverse NUP98-fusion proteins, we developed mouse models for regulatable expression of NUP98/NSD1, NUP98/JARID1A, and NUP98/DDX10. By integrating chromatin occupancy profiles of NUP98-fusion proteins with transcriptome profiling upon acute fusion protein inactivation in vivo, we defined the core set of direct transcriptional targets of NUP98-fusion proteins. Among those, CDK6 was highly expressed in murine and human AML samples. Loss of CDK6 severely attenuated NUP98-fusion–driven leukemogenesis, and NUP98-fusion AML was sensitive to pharmacologic CDK6 inhibition in vitro and in vivo. These findings identify CDK6 as a conserved, critical direct target of NUP98-fusion proteins, proposing CDK4/CDK6 inhibitors as a new rational treatment option for AML patients with NUP98-fusions.
- Published
- 2019
46. Screening of clustered regulatory elements reveals functional cooperating dependencies in Leukemia
- Author
-
Marion Antonini, Zakia Aid, Bryan Pardieu, Séverine Lecourt, Marie-Charlotte Laiguillon, Cécile Thirant, Arnaud Petit, Cécile K. Lopez, Eralda Salataj, Camille Lobry, Alexandre Puissant, Thomas Mercher, Julie Chaumeil, and Salima Benbarche
- Subjects
BRD4 ,Massive parallel sequencing ,Regulatory sequence ,medicine ,Cancer ,Computational biology ,Biology ,medicine.disease ,Genome ,Gene ,Fusion protein ,Epigenomics - Abstract
In the recent years, massively parallel sequencing approaches identified hundreds of mutated genes in cancer(1) providing an unprecedented amount of information about mechanisms of cancer cell maintenance and progression. However, while (it is widely accepted that) transformation processes result from oncogenic cooperation between deregulated genes and pathways, the functional characterization of candidate key players is mostly performed at the single gene level which is generally inadequate to identify these oncogene circuitries. In addition, studies aimed at depicting oncogenic cooperation involve the generation of challenging mouse models or the deployment of tedious screening pipelines. Genome wide mapping of epigenomic modifications on histone tails or binding of factors such as MED1 and BRD4 allowed identification of clusters of regulatory elements, also termed Super-Enhancers (SE)(2). Functional annotation of these regions revealed their high relevance during normal tissue development and cancer ontogeny(3). An interesting paradigm of the tumorigenic function of these SE regions comes from ETO2-GLIS2-driven acute megakaryoblastic leukemia (AMKL) in which the fusion protein ETO2-GLIS2 is sufficient to promote an aberrant transcriptional network by the rewiring of SE regions(4). We thus hypothesized that important regulatory regions could control simultaneously expression of genes cooperating in functional modules to promote cancer development. In an effort to identify such modules, we deployed a genome-wide CRISPRi-based screening approach and nominated SE regions that are functionally linked to leukemia maintenance. In particular, we pinpointed a novel SE region regulating the expression of both tyrosine kinases KIT and PDGFRA. Whereas the inhibition of each kinase alone affected modestly cancer cell growth, combined inhibition of both receptors synergizes to impair leukemia cell growth and survival. Our results demonstrate that genome-wide screening of regulatory DNA elements can identify co-regulated genes collaborating to promote cancer and could open new avenues to the concept of combined gene inhibition upon single hit targeting.
- Published
- 2019
- Full Text
- View/download PDF
47. Trials in Progress: A Phase I Study to Evaluate the Safety and Pharmacokinetic Profiles of CB-5339 in Participants with Relapsed/Refractory Acute Myeloid Leukemia or Relapsed/Refractory Intermediate or High-Risk Myelodysplastic Syndrome
- Author
-
Eytan M. Stein, Amy Burroughs, Monic J. Stuart, Lina Benajiba, Nada Hamad, Courtney D. DiNardo, Hetty E. Carraway, Costas K. Yannakou, Jesse D. Vargas, Scott Harris, Kimberly Stegmaier, Dorothy Nguyen, Hayley Lane, and Alexandre Puissant
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Immunology ,Myeloid leukemia ,Cell Biology ,Hematology ,Biochemistry ,Phase i study ,Pharmacokinetics ,Internal medicine ,Relapsed refractory ,medicine ,business - Abstract
Background: Acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) are clonal hematopoietic disorders that predominantly occur in older adults. For a limited number of fit patients, induction followed by consolidation chemotherapy and/or allogeneic stem-cell transplant (SCT) leads to cure. However, many AML patients are ineligible for aggressive therapy; up to 30% have primary refractory disease and up to 50% will relapse after front-line therapy, requiring alternative approaches. For these patients, durable long-term remission rate is low, with 5-year overall survival rates lower than 10%. Therefore, there remains an urgent unmet need for novel therapies for such patients. CB-5339 is a second generation, potent and selective, orally bioavailable small molecule inhibitor of valosin containing protein (VCP)/p97. VCP/p97 is a key cellular enzyme involved in cellular stress response pathways critical to cancer cell growth and survival such as protein homeostasis and the response to DNA damage. Inhibition of VCP/p97 in a panel of 131 cancer cell lines representing 16 cancer types revealed AML as the most exquisitely dependent disease (p=0.004) on VCP/p97 function. Further, CB-5339 demonstrated antiproliferative potency in a panel of 16 AML cell lines (IC50:100nM - 500nM). To better characterize the effects of CB-5339 on human leukemia, viability assays were performed on a set of 30 genetically diverse primary AML patient samples. Cellular viability was impacted with a similar potency across samples, irrespective of underlying genetic abnormalities (mean IC50: 423nM). In vivo, CB-5339 treatment resulted in decreased circulating leukemic cells and significantly prolonged survival in an MLL-AF9 syngeneic AML mouse model (p=0.02). In addition, evidence of synergy was exhibited with standard of care AML therapy, a combination of an anthracycline and cytarabine. This triple combination regimen resulted in a 96% mean relative decrease in leukemic burden compared to control mice and significantly prolonged mice survival compared to each regimen alone (p Here, we present a phase 1 study to evaluate CB-5339 in patients with relapsed/refractory (R/R) AML and intermediate or higher-risk MDS. Study design: This is a single arm, open-label, multi-center phase 1 clinical trial. The study includes two parts: 1) a Dose Escalation phase using a single-participant cohort accelerated titration schema in R/R AML or intermediate/higher-risk MDS (IPSS-R) participants and 2) a Dose Expansion phase using a 2 stage design for further assessment of disease specific cohorts and potential combination strategies at the recommended phase 2 dose (RP2D). CB-5339 will be administered orally once-daily (QD) for 4 days, followed by a 3-day treatment-free period weekly, in successive 28-day cycles until progressive disease or intolerable toxicity. Intraparticipant dose escalation will be permitted after Cycle 1. Twice-daily (BID) dosing may also be tested depending on initial PK assessments. The primary objective of this trial is to determine the safety, tolerability, and MTD and/or RP2D of CB-5339. The secondary objectives are to characterize PK properties and antitumor activity of CB-5339. Exploratory objectives will evaluate potential pharmacodynamic (PD) biomarkers and characterize the PK/PD relationship. Key inclusion criteria include participants with pathologically confirmed AML or intermediate/higher-risk MDS relapsed and/or refractory to standard therapies, ECOG ≤2, and adequate kidney and liver functions. Key exclusion criteria include pathologically confirmed acute promyelocytic leukemia, central nervous system involvement, immediately life-threatening/severe complications of AML/MDS such as uncontrolled bleeding, uncontrolled active infection, pneumonia with hypoxia or shock, and/or disseminated intravascular coagulation, SCT within 100 days of first dose, active GVHD, or post-SCT systemic immunosuppressive therapy, concomitant active malignancy requiring chemotherapy and adverse cardiac history. Up to 60 participants (20 in dose escalation and 40 in dose expansion) will be enrolled at approximately 10 sites in the U.S and Australia. Recruitment is ongoing and this trial is registered with clinicaltrials.gov: NCT04402541. Disclosures Benajiba: Gilead Foundation: Research Funding. Carraway:BMS: Consultancy, Other: Research support, Speakers Bureau; Novartis: Consultancy, Speakers Bureau; Jazz: Consultancy, Speakers Bureau; Stemline: Consultancy, Speakers Bureau; Takeda: Other: Independent Advisory Committe (IRC); ASTEX: Other: Independent Advisory Committe (IRC); Abbvie: Other: Independent Advisory Committe (IRC). Hamad:Novartis: Honoraria; Abbvie: Honoraria. Stein:Biotheryx: Consultancy; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; PTC Therapeutics: Membership on an entity's Board of Directors or advisory committees; Syros: Membership on an entity's Board of Directors or advisory committees; Syndax: Consultancy, Research Funding; Genentech: Consultancy, Membership on an entity's Board of Directors or advisory committees; Bayer: Research Funding; Seattle Genetics: Consultancy; Daiichi-Sankyo: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Consultancy; Astellas Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees; Agios Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celgene Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Abbvie: Consultancy. Burroughs:Chiasma: Current equity holder in publicly-traded company; Synthetic Genomics: Current equity holder in private company; Cleave Therapeutics: Current Employment, Current equity holder in private company; Crinetics Pharmaceuticals: Current equity holder in publicly-traded company, Ended employment in the past 24 months. Harris:Cleave Therapeutics: Current Employment, Current equity holder in private company. Lane:Cleave Therapeutics: Consultancy; Regimmune Corporation: Consultancy; Star Therapeutics: Consultancy; Nanoscope Therapeutics: Consultancy; Viewpoint Therapeutics: Ended employment in the past 24 months; Valitor: Consultancy; Revolution Medicines: Consultancy. Nguyen:Cleave Therapeutics, Inc: Current Employment, Current equity holder in private company. Stuart:Cleave Therapeutics: Consultancy; Triphase Accelerator U.S Corporation: Consultancy; Revolution Medicines Inc: Consultancy; RegImmune Corp: Consultancy; IgM Biosciences: Consultancy; Portola Pharmaceuticals Inc: Consultancy; Artiva Biotherapeutics: Consultancy; Gilead Sciences Inc: Consultancy; Theravance Biopharma: Consultancy; Integral Medicines Inc: Consultancy. Vargas:Cytomx Therapeutics, Inc.: Current equity holder in publicly-traded company; Sangamo Therapeutics: Current equity holder in publicly-traded company; Cleave Therapeutics, Inc.: Current Employment, Current equity holder in private company. Stegmaier:Novartis: Research Funding; Auron Therapeutics: Consultancy. DiNardo:ImmuneOnc: Honoraria; Celgene: Consultancy, Honoraria, Research Funding; Daiichi Sankyo: Consultancy, Honoraria, Research Funding; Takeda: Honoraria; Notable Labs: Membership on an entity's Board of Directors or advisory committees; AbbVie: Consultancy, Honoraria, Research Funding; MedImmune: Honoraria; Agios: Consultancy, Honoraria, Research Funding; Jazz: Honoraria; Novartis: Consultancy; Calithera: Research Funding; Syros: Honoraria.
- Published
- 2020
- Full Text
- View/download PDF
48. Creatine kinase pathway inhibition alters GSK3 and WNT signaling in EVI1-positive AML
- Author
-
Olivier Hermine, Kimberly Stegmaier, Emmanuel Raffoux, Lina Benajiba, Raphael Itzykson, Jean Soulier, Alexandre Puissant, Angela Su, Gabriela Alexe, and Michael T. Hemann
- Subjects
0301 basic medicine ,Cancer Research ,Myeloid ,Article ,Glycogen Synthase Kinase 3 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Text mining ,Animals ,Humans ,Medicine ,Creatine Kinase ,Wnt Signaling Pathway ,biology ,Extramural ,business.industry ,Wnt signaling pathway ,Hematology ,medicine.disease ,MDS1 and EVI1 Complex Locus Protein ,Wnt Proteins ,Leukemia, Myeloid, Acute ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Creatine kinase ,business - Published
- 2019
- Full Text
- View/download PDF
49. LAMP2 expression dictates azacytidine response and prognosis in MDS/AML
- Author
-
Alexandre Puissant, François Orange, Guillaume Robert, Marwa Zerhouni, Thomas Cluzeau, Mohamed Amine Hamouda, Jean-Michel Karsenti, Frederic Luciano, Nathan Furstoss, Coline Savy, Jerome Tamburini, Arnaud Jacquel, Nicolas Mounier, Patrick Auberger, Sandrine Marchetti, Sandra Lacas-Gervais, Alix Dubois, Anne Calleja, and Sonia Boulakirba
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Antimetabolites, Antineoplastic ,Myeloid ,CD34 ,Apoptosis ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Lysosome ,Lysosomal-Associated Membrane Protein 2 ,medicine ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Humans ,Chemotherapy ,Receptor ,neoplasms ,Loss function ,Aged ,Cell Proliferation ,Aged, 80 and over ,LAMP2 ,Cell growth ,business.industry ,Autophagy ,Hematology ,Middle Aged ,medicine.disease ,Prognosis ,Gene Expression Regulation, Neoplastic ,Survival Rate ,Leukemia ,Leukemia, Myeloid, Acute ,030104 developmental biology ,medicine.anatomical_structure ,Retraction Note ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,Azacitidine ,Female ,Bone marrow ,business ,Myelodysplastic syndrome ,Follow-Up Studies - Abstract
Chaperone-mediated autophagy (CMA) is a highly selective form of autophagy. During CMA, the HSC70 chaperone carries target proteins endowed with a KFERQ-like motif to the lysosomal receptor LAMP2A, which then translocate them into lysosomes for degradation. In the present study, we scrutinized the mechanisms underlying the response and resistance to Azacytidine (Aza) in MDS/AML cell lines and bone marrow CD34+ blasts from MDS/AML patients. In engineered Aza-resistant MDS cell lines and some AML cell lines, we identified a profound defect in CMA linked to the absence of LAMP2A. LAMP2 deficiency was responsible for Aza resistance and hypersensitivity to lysosome and autophagy inhibitors. Accordingly, gain of function of LAMP2 in deficient cells or loss of function in LAMP2-expressing cells rendered them sensitive or resistant to Aza, respectively. A strict correlation was observed between the absence of LAMP2, resistance to Aza and sensitivity to lysosome inhibitors. Low levels of LAMP2 expression in CD34+ blasts from MDS/AML patients correlated with lack of sensitivity to Aza and were predictive of poor overall survival. We propose that CD34+/LAMP2Low patients at diagnosis or who become CD34+/LAMP2Low during the course of treatment with Aza might benefit from a lysosome inhibitor already used in the clinic.
- Published
- 2018
50. Exploiting an Asp-Glu 'switch' in glycogen synthase kinase 3 to design paralog-selective inhibitors for use in acute myeloid leukemia
- Author
-
Morgan Back, Matthew B. Robers, Edward M. Scolnick, Adam J. Stein, Jennifer Gale, Lina Benajiba, Damodharan Lakshminarasimhan, Arthur Campbell, Thomas Machleidt, Richard Stone, Florence F. Wagner, Xi Shi, Michael T. Hemann, Jennifer Wilkinson, Amy Saur Conway, Andrew L. Kung, Edward Holson, Joshua R. Sacher, Alexandre Puissant, Yan-Ling Zhang, Daniel J. DeAngelo, Yana Pikman, Ilene Galinsky, Linda Ross, Jen Q. Pan, Olivier Hermine, Kimberly Stegmaier, Gabriela Alexe, Taner Kaya, Michel Weiwer, Université Paris-Saclay, Laboratoire de chimie des Molécules bioactives et des aromes ( cnrs, umr 6001 ), Université Nice Sophia Antipolis ( UNS ), Université Côte d'Azur ( UCA ) -Université Côte d'Azur ( UCA ), Dana-Farber Cancer Institute [Boston], Computational Biology Center ( IBM T.J. Watson Research Center ), IBM, Centre de référence des mastocytoses, CHU Necker - Enfants Malades [AP-HP], Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, and Department of Pediatric Oncology [Boston, MA, USA]
- Subjects
0301 basic medicine ,Myeloid ,animal structures ,Biology ,Article ,[ SDV.CAN ] Life Sciences [q-bio]/Cancer ,Glycogen Synthase Kinase 3 ,03 medical and health sciences ,GSK-3 ,medicine ,Humans ,Protein Isoforms ,Enzyme Inhibitors ,beta Catenin ,Drug discovery ,Kinase ,Wnt signaling pathway ,Myeloid leukemia ,Dipeptides ,U937 Cells ,General Medicine ,medicine.disease ,3. Good health ,Leukemia, Myeloid, Acute ,Leukemia ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Mutagenesis, Site-Directed ,Cancer research - Abstract
International audience; Glycogen synthase kinase 3 (GSK3), a key regulatory kinase in the wingless-type MMTV integration site family (WNT) pathway, is a therapeutic target of interest in many diseases. Although dual GSK3α/β inhibitors have entered clinical trials, none has successfully translated to clinical application. Mechanism-based toxicities, driven in part by the inhibition of both GSK3 paralogs and subsequent β-catenin stabilization, are a concern in the translation of this target class because mutations and overexpression of β-catenin are associated with many cancers. Knockdown of GSK3α or GSK3β individually does not increase β-catenin and offers a conceptual resolution to targeting GSK3: paralog-selective inhibition. However, inadequate chemical tools exist. The design of selective adenosine triphosphate (ATP)-competitive inhibitors poses a drug discovery challenge due to the high homology (95% identity and 100% similarity) in this binding domain. Taking advantage of an Asp133→Glu196 "switch" in their kinase hinge, we present a rational design strategy toward the discovery of paralog-selective GSK3 inhibitors. These GSK3α- and GSK3β-selective inhibitors provide insights into GSK3 targeting in acute myeloid leukemia (AML), where GSK3α was identified as a therapeutic target using genetic approaches. The GSK3α-selective compound BRD0705 inhibits kinase function and does not stabilize β-catenin, mitigating potential neoplastic concerns. BRD0705 induces myeloid differentiation and impairs colony formation in AML cells, with no apparent effect on normal hematopoietic cells. Moreover, BRD0705 impairs leukemia initiation and prolongs survival in AML mouse models. These studies demonstrate feasibility of paralog-selective GSK3α inhibition, offering a promising therapeutic approach in AML.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.