Skip to main content
Top
Published in: Annals of Hematology 12/2022

05-10-2022 | Erythropoietin | Original Article

Luspatercept (RAP-536) modulates oxidative stress without affecting mutation burden in myelodysplastic syndromes

Authors: Meunier Mathieu, Chloé Friedrich, Nicolas Ducrot, Johanna Zannoni, Tondeur Sylvie, Nelly Jerraya, Sophie Rousseaux, Florent Chuffart, Olivier Kosmider, Zoubida Karim, Sophie Park

Published in: Annals of Hematology | Issue 12/2022

Login to get access

Abstract

In low-risk myelodysplastic syndrome (LR-MDS), erythropoietin (EPO) is widely used for the treatment of chronic anemia. However, initial response to EPO has time-limited effects. Luspatercept reduces red blood cell transfusion dependence in LR-MDS patients. Here, we investigated the molecular action of luspatercept (RAP-536) in an in vitro model of erythroid differentiation of MDS, and also in a in vivo PDX murine model with primary samples of MDS patients carrying or not SF3B1 mutation. In our in vitro model, RAP-536 promotes erythroid proliferation by increasing the number of cycling cells without any impact on apoptosis rates. RAP-536 promoted late erythroid precursor maturation while decreasing intracellular reactive oxygen species level. RNA sequencing of erythroid progenitors obtained under RAP-536 treatment showed an enrichment of genes implicated in positive regulation of response to oxidative stress and erythroid differentiation. In our PDX model, RAP-536 induces a higher hemoglobin level. RAP-536 did not modify variant allele frequencies in vitro and did not have any effect against leukemic burden in our PDX model. These results suggest that RAP-536 promotes in vivo and in vitro erythroid cell differentiation by decreasing ROS level without any remarkable impact on iron homeostasis and on mutated allele burden.
Appendix
Available only for authorised users
Literature
1.
go back to reference Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, Bloomfield CD, Cazzola M, Vardiman JW (2016) The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 127:2391–2405CrossRefPubMed Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, Bloomfield CD, Cazzola M, Vardiman JW (2016) The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 127:2391–2405CrossRefPubMed
2.
go back to reference Zeidan AM, Shallis RM, Wang R, Davidoff A, Ma X (2019) Epidemiology of myelodysplastic syndromes: why characterizing the beast is a prerequisite to taming it. Blood Rev 34:1–15CrossRefPubMed Zeidan AM, Shallis RM, Wang R, Davidoff A, Ma X (2019) Epidemiology of myelodysplastic syndromes: why characterizing the beast is a prerequisite to taming it. Blood Rev 34:1–15CrossRefPubMed
3.
go back to reference Greenberg P, Cox C, LeBeau MM, Fenaux P, Morel P, Sanz G, Sanz M, Vallespi T, Hamblin T, Oscier D, Ohyashiki K, Toyama K, Aul C, Mufti G, Bennett J (1997) International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood 89:2079–2088CrossRefPubMed Greenberg P, Cox C, LeBeau MM, Fenaux P, Morel P, Sanz G, Sanz M, Vallespi T, Hamblin T, Oscier D, Ohyashiki K, Toyama K, Aul C, Mufti G, Bennett J (1997) International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood 89:2079–2088CrossRefPubMed
4.
go back to reference Greenberg PL, Tuechler H, Schanz J, Sanz G, Garcia-Manero G, Sole F, Bennett JM, Bowen D, Fenaux P, Dreyfus F, Kantarjian H, Kuendgen A, Levis A, Malcovati L, Cazzola M, Cermak J, Fonatsch C, Le Beau MM, Slovak ML, Krieger O, Luebbert M, Maciejewski J, Magalhaes SM, Miyazaki Y, Pfeilstocker M, Sekeres M, Sperr WR, Stauder R, Tauro S, Valent P, Vallespi T, van de Loosdrecht AA, Germing U, Haase D (2012) Revised international prognostic scoring system for myelodysplastic syndromes. Blood 120:2454–2465CrossRefPubMedPubMedCentral Greenberg PL, Tuechler H, Schanz J, Sanz G, Garcia-Manero G, Sole F, Bennett JM, Bowen D, Fenaux P, Dreyfus F, Kantarjian H, Kuendgen A, Levis A, Malcovati L, Cazzola M, Cermak J, Fonatsch C, Le Beau MM, Slovak ML, Krieger O, Luebbert M, Maciejewski J, Magalhaes SM, Miyazaki Y, Pfeilstocker M, Sekeres M, Sperr WR, Stauder R, Tauro S, Valent P, Vallespi T, van de Loosdrecht AA, Germing U, Haase D (2012) Revised international prognostic scoring system for myelodysplastic syndromes. Blood 120:2454–2465CrossRefPubMedPubMedCentral
5.
go back to reference Santini V (2015) Anemia as the main manifestation of myelodysplastic syndromes. Semin Hematol 52:348–356CrossRefPubMed Santini V (2015) Anemia as the main manifestation of myelodysplastic syndromes. Semin Hematol 52:348–356CrossRefPubMed
6.
go back to reference Sekeres MA (2011) Epidemiology, natural history, and practice patterns of patients with myelodysplastic syndromes in 2010. J Natl Compr Canc Netw 9:57–63CrossRefPubMed Sekeres MA (2011) Epidemiology, natural history, and practice patterns of patients with myelodysplastic syndromes in 2010. J Natl Compr Canc Netw 9:57–63CrossRefPubMed
7.
go back to reference Balducci L (2006) Transfusion independence in patients with myelodysplastic syndromes: impact on outcomes and quality of life. Cancer 106:2087–2094CrossRefPubMed Balducci L (2006) Transfusion independence in patients with myelodysplastic syndromes: impact on outcomes and quality of life. Cancer 106:2087–2094CrossRefPubMed
8.
go back to reference Stauder R, Yu G, Koinig KA, Bagguley T, Fenaux P, Symeonidis A, Sanz G, Cermak J, Mittelman M, Hellstrom-Lindberg E, Langemeijer S, Holm MS, Madry K, Malcovati L, Tatic A, Germing U, Savic A, van Marrewijk C, Guerci-Bresler A, Luno E, Droste J, Efficace F, Smith A, Bowen D, de Witte T (2018) Health-related quality of life in lower-risk MDS patients compared with age- and sex-matched reference populations: a European LeukemiaNet study. Leukemia 32:1380–1392CrossRefPubMedPubMedCentral Stauder R, Yu G, Koinig KA, Bagguley T, Fenaux P, Symeonidis A, Sanz G, Cermak J, Mittelman M, Hellstrom-Lindberg E, Langemeijer S, Holm MS, Madry K, Malcovati L, Tatic A, Germing U, Savic A, van Marrewijk C, Guerci-Bresler A, Luno E, Droste J, Efficace F, Smith A, Bowen D, de Witte T (2018) Health-related quality of life in lower-risk MDS patients compared with age- and sex-matched reference populations: a European LeukemiaNet study. Leukemia 32:1380–1392CrossRefPubMedPubMedCentral
9.
go back to reference Moyo V, Lefebvre P, Duh MS, Yektashenas B, Mundle S (2008) Erythropoiesis-stimulating agents in the treatment of anemia in myelodysplastic syndromes: a meta-analysis. Ann Hematol 87:527–536CrossRefPubMed Moyo V, Lefebvre P, Duh MS, Yektashenas B, Mundle S (2008) Erythropoiesis-stimulating agents in the treatment of anemia in myelodysplastic syndromes: a meta-analysis. Ann Hematol 87:527–536CrossRefPubMed
10.
go back to reference Park S, Hamel JF, Toma A, Kelaidi C, Thepot S, Campelo MD, Santini V, Sekeres MA, Balleari E, Kaivers J, Sapena R, Gotze K, Muller-Thomas C, Beyne-Rauzy O, Stamatoullas A, Kotsianidis I, Komrokji R, Steensma DP, Fensterl J, Roboz GJ, Bernal T, Ramos F, Calabuig M, Guerci-Bresler A, Bordessoule D, Cony-Makhoul P, Cheze S, Wattel E, Rose C, Vey N, Gioia D, Ferrero D, Gaidano G, Cametti G, Pane F, Sanna A, Germing U, Sanz GF, Dreyfus F, Fenaux P (2017) Outcome of lower-risk patients with myelodysplastic syndromes without 5q deletion after failure of erythropoiesis-stimulating agents. J Clin Oncol 35:1591–1597CrossRefPubMed Park S, Hamel JF, Toma A, Kelaidi C, Thepot S, Campelo MD, Santini V, Sekeres MA, Balleari E, Kaivers J, Sapena R, Gotze K, Muller-Thomas C, Beyne-Rauzy O, Stamatoullas A, Kotsianidis I, Komrokji R, Steensma DP, Fensterl J, Roboz GJ, Bernal T, Ramos F, Calabuig M, Guerci-Bresler A, Bordessoule D, Cony-Makhoul P, Cheze S, Wattel E, Rose C, Vey N, Gioia D, Ferrero D, Gaidano G, Cametti G, Pane F, Sanna A, Germing U, Sanz GF, Dreyfus F, Fenaux P (2017) Outcome of lower-risk patients with myelodysplastic syndromes without 5q deletion after failure of erythropoiesis-stimulating agents. J Clin Oncol 35:1591–1597CrossRefPubMed
11.
go back to reference Zermati Y, Fichelson S, Valensi F, Freyssinier JM, Rouyer-Fessard P, Cramer E, Guichard J, Varet B, Hermine O (2000) Transforming growth factor inhibits erythropoiesis by blocking proliferation and accelerating differentiation of erythroid progenitors. Exp Hematol 28:885–894CrossRefPubMed Zermati Y, Fichelson S, Valensi F, Freyssinier JM, Rouyer-Fessard P, Cramer E, Guichard J, Varet B, Hermine O (2000) Transforming growth factor inhibits erythropoiesis by blocking proliferation and accelerating differentiation of erythroid progenitors. Exp Hematol 28:885–894CrossRefPubMed
12.
go back to reference Antebi YE, Linton JM, Klumpe H, Bintu B, Gong M, Su C, McCardell R, Elowitz MB (2017) Combinatorial signal perception in the BMP pathway. Cell 170:1184-1196.e1124CrossRefPubMedPubMedCentral Antebi YE, Linton JM, Klumpe H, Bintu B, Gong M, Su C, McCardell R, Elowitz MB (2017) Combinatorial signal perception in the BMP pathway. Cell 170:1184-1196.e1124CrossRefPubMedPubMedCentral
13.
go back to reference Neuzillet C, Tijeras-Raballand A, Cohen R, Cros J, Faivre S, Raymond E, de Gramont A (2015) Targeting the TGFbeta pathway for cancer therapy. Pharmacol Ther 147:22–31CrossRefPubMed Neuzillet C, Tijeras-Raballand A, Cohen R, Cros J, Faivre S, Raymond E, de Gramont A (2015) Targeting the TGFbeta pathway for cancer therapy. Pharmacol Ther 147:22–31CrossRefPubMed
14.
go back to reference Soderberg SS, Karlsson G, Karlsson S (2009) Complex and context dependent regulation of hematopoiesis by TGF-beta superfamily signaling. Ann N Y Acad Sci 1176:55–69CrossRefPubMed Soderberg SS, Karlsson G, Karlsson S (2009) Complex and context dependent regulation of hematopoiesis by TGF-beta superfamily signaling. Ann N Y Acad Sci 1176:55–69CrossRefPubMed
15.
go back to reference Blank U, Karlsson S (2011) The role of Smad signaling in hematopoiesis and translational hematology. Leukemia 25:1379–1388CrossRefPubMed Blank U, Karlsson S (2011) The role of Smad signaling in hematopoiesis and translational hematology. Leukemia 25:1379–1388CrossRefPubMed
16.
go back to reference Suragani RN, Cadena SM, Cawley SM, Sako D, Mitchell D, Li R, Davies MV, Alexander MJ, Devine M, Loveday KS, Underwood KW, Grinberg AV, Quisel JD, Chopra R, Pearsall RS, Seehra J, Kumar R (2014) Transforming growth factor-beta superfamily ligand trap ACE-536 corrects anemia by promoting late-stage erythropoiesis. Nat Med 20:408–414CrossRefPubMed Suragani RN, Cadena SM, Cawley SM, Sako D, Mitchell D, Li R, Davies MV, Alexander MJ, Devine M, Loveday KS, Underwood KW, Grinberg AV, Quisel JD, Chopra R, Pearsall RS, Seehra J, Kumar R (2014) Transforming growth factor-beta superfamily ligand trap ACE-536 corrects anemia by promoting late-stage erythropoiesis. Nat Med 20:408–414CrossRefPubMed
17.
go back to reference Zhou L, McMahon C, Bhagat T, Alencar C, Yu Y, Fazzari M, Sohal D, Heuck C, Gundabolu K, Ng C, Mo Y, Shen W, Wickrema A, Kong G, Friedman E, Sokol L, Mantzaris I, Pellagatti A, Boultwood J, Platanias LC, Steidl U, Yan L, Yingling JM, Lahn MM, List A, Bitzer M, Verma A (2011) Reduced SMAD7 leads to overactivation of TGF-beta signaling in MDS that can be reversed by a specific inhibitor of TGF-beta receptor I kinase. Cancer Res 71:955–963CrossRefPubMed Zhou L, McMahon C, Bhagat T, Alencar C, Yu Y, Fazzari M, Sohal D, Heuck C, Gundabolu K, Ng C, Mo Y, Shen W, Wickrema A, Kong G, Friedman E, Sokol L, Mantzaris I, Pellagatti A, Boultwood J, Platanias LC, Steidl U, Yan L, Yingling JM, Lahn MM, List A, Bitzer M, Verma A (2011) Reduced SMAD7 leads to overactivation of TGF-beta signaling in MDS that can be reversed by a specific inhibitor of TGF-beta receptor I kinase. Cancer Res 71:955–963CrossRefPubMed
18.
go back to reference Arlet JB, Guillem F, Lamarque M, Dussiot M, Maciel T, Moura I, Hermine O, Courtois G (2016) Protein-based therapeutic for anemia caused by dyserythropoiesis. Expert Rev Proteomics 13:983–992CrossRefPubMed Arlet JB, Guillem F, Lamarque M, Dussiot M, Maciel T, Moura I, Hermine O, Courtois G (2016) Protein-based therapeutic for anemia caused by dyserythropoiesis. Expert Rev Proteomics 13:983–992CrossRefPubMed
19.
go back to reference Attie KM, Allison MJ, McClure T, Boyd IE, Wilson DM, Pearsall AE, Sherman ML (2014) A phase 1 study of ACE-536, a regulator of erythroid differentiation, in healthy volunteers. Am J Hematol 89:766–770CrossRefPubMedPubMedCentral Attie KM, Allison MJ, McClure T, Boyd IE, Wilson DM, Pearsall AE, Sherman ML (2014) A phase 1 study of ACE-536, a regulator of erythroid differentiation, in healthy volunteers. Am J Hematol 89:766–770CrossRefPubMedPubMedCentral
20.
go back to reference Platzbecker U, Germing U, Gotze KS, Kiewe P, Mayer K, Chromik J, Radsak M, Wolff T, Zhang X, Laadem A, Sherman ML, Attie KM, Giagounidis A (2017) Luspatercept for the treatment of anaemia in patients with lower-risk myelodysplastic syndromes (PACE-MDS): a multicentre, open-label phase 2 dose-finding study with long-term extension study. Lancet Oncol 18:1338–1347CrossRefPubMed Platzbecker U, Germing U, Gotze KS, Kiewe P, Mayer K, Chromik J, Radsak M, Wolff T, Zhang X, Laadem A, Sherman ML, Attie KM, Giagounidis A (2017) Luspatercept for the treatment of anaemia in patients with lower-risk myelodysplastic syndromes (PACE-MDS): a multicentre, open-label phase 2 dose-finding study with long-term extension study. Lancet Oncol 18:1338–1347CrossRefPubMed
21.
go back to reference Fenaux P, Platzbecker U, Mufti GJ, Garcia-Manero G, Buckstein R, Santini V, Diez-Campelo M, Finelli C, Cazzola M, Ilhan O, Sekeres MA, Falantes JF, Arrizabalaga B, Salvi F, Giai V, Vyas P, Bowen D, Selleslag D, DeZern AE, Jurcic JG, Germing U, Gotze KS, Quesnel B, Beyne-Rauzy O, Cluzeau T, Voso MT, Mazure D, Vellenga E, Greenberg PL, Hellstrom-Lindberg E, Zeidan AM, Ades L, Verma A, Savona MR, Laadem A, Benzohra A, Zhang J, Rampersad A, Dunshee DR, Linde PG, Sherman ML, Komrokji RS, List AF (2020) Luspatercept in patients with lower-risk myelodysplastic syndromes. N Engl J Med 382:140–151CrossRefPubMed Fenaux P, Platzbecker U, Mufti GJ, Garcia-Manero G, Buckstein R, Santini V, Diez-Campelo M, Finelli C, Cazzola M, Ilhan O, Sekeres MA, Falantes JF, Arrizabalaga B, Salvi F, Giai V, Vyas P, Bowen D, Selleslag D, DeZern AE, Jurcic JG, Germing U, Gotze KS, Quesnel B, Beyne-Rauzy O, Cluzeau T, Voso MT, Mazure D, Vellenga E, Greenberg PL, Hellstrom-Lindberg E, Zeidan AM, Ades L, Verma A, Savona MR, Laadem A, Benzohra A, Zhang J, Rampersad A, Dunshee DR, Linde PG, Sherman ML, Komrokji RS, List AF (2020) Luspatercept in patients with lower-risk myelodysplastic syndromes. N Engl J Med 382:140–151CrossRefPubMed
22.
go back to reference Wobus M, Mies A, Asokan N, Oelschlagel U, Mobus K, Winter S, Cross M, Weidner H, Rauner M, Hofbauer LC, Bornhauser M, Platzbecker U (2021) Luspatercept restores SDF-1-mediated hematopoietic support by MDS-derived mesenchymal stromal cells. Leukemia 35:2936–2947CrossRefPubMedPubMedCentral Wobus M, Mies A, Asokan N, Oelschlagel U, Mobus K, Winter S, Cross M, Weidner H, Rauner M, Hofbauer LC, Bornhauser M, Platzbecker U (2021) Luspatercept restores SDF-1-mediated hematopoietic support by MDS-derived mesenchymal stromal cells. Leukemia 35:2936–2947CrossRefPubMedPubMedCentral
23.
go back to reference Hu J, Liu J, Xue F, Halverson G, Reid M, Guo A, Chen L, Raza A, Galili N, Jaffray J, Lane J, Chasis JA, Taylor N, Mohandas N, An X (2013) Isolation and functional characterization of human erythroblasts at distinct stages: implications for understanding of normal and disordered erythropoiesis in vivo. Blood 121:3246–3253CrossRefPubMedPubMedCentral Hu J, Liu J, Xue F, Halverson G, Reid M, Guo A, Chen L, Raza A, Galili N, Jaffray J, Lane J, Chasis JA, Taylor N, Mohandas N, An X (2013) Isolation and functional characterization of human erythroblasts at distinct stages: implications for understanding of normal and disordered erythropoiesis in vivo. Blood 121:3246–3253CrossRefPubMedPubMedCentral
24.
go back to reference Meunier M, Ancelet S, Lefebvre C, Arnaud J, Garrel C, Pezet M, Wang Y, Faure P, Szymanski G, Duployez N, Preudhomme C, Biard D, Polack B, Cahn JY, Moulis JM, Park S (2017) Reactive oxygen species levels control NF-kappaB activation by low dose deferasirox in erythroid progenitors of low risk myelodysplastic syndromes. Oncotarget 8:105510–105524CrossRefPubMedPubMedCentral Meunier M, Ancelet S, Lefebvre C, Arnaud J, Garrel C, Pezet M, Wang Y, Faure P, Szymanski G, Duployez N, Preudhomme C, Biard D, Polack B, Cahn JY, Moulis JM, Park S (2017) Reactive oxygen species levels control NF-kappaB activation by low dose deferasirox in erythroid progenitors of low risk myelodysplastic syndromes. Oncotarget 8:105510–105524CrossRefPubMedPubMedCentral
25.
go back to reference Zheng QQ, Zhao YS, Guo J, Zhao SD, Song LX, Fei CM, Zhang Z, Li X, Chang CK (2017) Iron overload promotes erythroid apoptosis through regulating HIF-1a/ROS signaling pathway in patients with myelodysplastic syndrome. Leuk Res 58:55–62CrossRefPubMed Zheng QQ, Zhao YS, Guo J, Zhao SD, Song LX, Fei CM, Zhang Z, Li X, Chang CK (2017) Iron overload promotes erythroid apoptosis through regulating HIF-1a/ROS signaling pathway in patients with myelodysplastic syndrome. Leuk Res 58:55–62CrossRefPubMed
26.
go back to reference Meunier M, Dussiau C, Mauz N, Alary AS, Lefebvre C, Szymanski G, Pezet M, Blanquet F, Kosmider O, Park S (2018) Molecular dissection of engraftment in a xenograft model of myelodysplastic syndromes. Oncotarget 9:14993–15000CrossRefPubMedPubMedCentral Meunier M, Dussiau C, Mauz N, Alary AS, Lefebvre C, Szymanski G, Pezet M, Blanquet F, Kosmider O, Park S (2018) Molecular dissection of engraftment in a xenograft model of myelodysplastic syndromes. Oncotarget 9:14993–15000CrossRefPubMedPubMedCentral
27.
go back to reference Chan LSA, Gu LC, Leitch HA, Wells RA (2021) Intracellular ROS profile in hematopoietic progenitors of MDS patients: association with blast count and iron overload. Hematology 26:88–95CrossRefPubMed Chan LSA, Gu LC, Leitch HA, Wells RA (2021) Intracellular ROS profile in hematopoietic progenitors of MDS patients: association with blast count and iron overload. Hematology 26:88–95CrossRefPubMed
28.
go back to reference Park S, Kosmider O, Maloisel F, Drenou B, Chapuis N, Lefebvre T, Karim Z, Puy H, Alary AS, Ducamp S, Verdier F, Bouilloux C, Rousseau A, Jacob MC, Debliquis A, Charpentier A, Gyan E, Anglaret B, Leyronnas C, Corm S, Slama B, Cheze S, Laribi K, Ame S, Rose C, Lachenal F, Toma A, Pica GM, Carre M, Garban F, Mariette C, Cahn JY, Meunier M, Herault O, Fenaux P, Wagner-Ballon O, Bardet V, Dreyfus F, Fontenay M (2019) Dyserythropoiesis evaluated by the RED score and hepcidin:ferritin ratio predicts response to erythropoietin in lower-risk myelodysplastic syndromes. Haematologica 104:497–504CrossRefPubMedPubMedCentral Park S, Kosmider O, Maloisel F, Drenou B, Chapuis N, Lefebvre T, Karim Z, Puy H, Alary AS, Ducamp S, Verdier F, Bouilloux C, Rousseau A, Jacob MC, Debliquis A, Charpentier A, Gyan E, Anglaret B, Leyronnas C, Corm S, Slama B, Cheze S, Laribi K, Ame S, Rose C, Lachenal F, Toma A, Pica GM, Carre M, Garban F, Mariette C, Cahn JY, Meunier M, Herault O, Fenaux P, Wagner-Ballon O, Bardet V, Dreyfus F, Fontenay M (2019) Dyserythropoiesis evaluated by the RED score and hepcidin:ferritin ratio predicts response to erythropoietin in lower-risk myelodysplastic syndromes. Haematologica 104:497–504CrossRefPubMedPubMedCentral
29.
30.
go back to reference Bouscary D, De Vos J, Guesnu M, Jondeau K, Viguier F, Melle J, Picard F, Dreyfus F, Fontenay-Roupie M (1997) Fas/Apo-1 (CD95) expression and apoptosis in patients with myelodysplastic syndromes. Leukemia 11:839–845CrossRefPubMed Bouscary D, De Vos J, Guesnu M, Jondeau K, Viguier F, Melle J, Picard F, Dreyfus F, Fontenay-Roupie M (1997) Fas/Apo-1 (CD95) expression and apoptosis in patients with myelodysplastic syndromes. Leukemia 11:839–845CrossRefPubMed
31.
go back to reference Claessens YE, Park S, Dubart-Kupperschmitt A, Mariot V, Garrido C, Chretien S, Dreyfus F, Lacombe C, Mayeux P, Fontenay M (2005) Rescue of early-stage myelodysplastic syndrome-deriving erythroid precursors by the ectopic expression of a dominant-negative form of FADD. Blood 105:4035–4042CrossRefPubMed Claessens YE, Park S, Dubart-Kupperschmitt A, Mariot V, Garrido C, Chretien S, Dreyfus F, Lacombe C, Mayeux P, Fontenay M (2005) Rescue of early-stage myelodysplastic syndrome-deriving erythroid precursors by the ectopic expression of a dominant-negative form of FADD. Blood 105:4035–4042CrossRefPubMed
32.
go back to reference Suragani RN, Cawley SM, Li R, Wallner S, Alexander MJ, Mulivor AW, Gardenghi S, Rivella S, Grinberg AV, Pearsall RS, Kumar R (2014) Modified activin receptor IIB ligand trap mitigates ineffective erythropoiesis and disease complications in murine beta-thalassemia. Blood 123:3864–3872CrossRefPubMedPubMedCentral Suragani RN, Cawley SM, Li R, Wallner S, Alexander MJ, Mulivor AW, Gardenghi S, Rivella S, Grinberg AV, Pearsall RS, Kumar R (2014) Modified activin receptor IIB ligand trap mitigates ineffective erythropoiesis and disease complications in murine beta-thalassemia. Blood 123:3864–3872CrossRefPubMedPubMedCentral
33.
go back to reference Chai X, Li D, Cao X, Zhang Y, Mu J, Lu W, Xiao X, Li C, Meng J, Chen J, Li Q, Wang J, Meng A, Zhao M (2015) ROS-mediated iron overload injures the hematopoiesis of bone marrow by damaging hematopoietic stem/progenitor cells in mice. Sci Rep 5:10181CrossRefPubMedPubMedCentral Chai X, Li D, Cao X, Zhang Y, Mu J, Lu W, Xiao X, Li C, Meng J, Chen J, Li Q, Wang J, Meng A, Zhao M (2015) ROS-mediated iron overload injures the hematopoiesis of bone marrow by damaging hematopoietic stem/progenitor cells in mice. Sci Rep 5:10181CrossRefPubMedPubMedCentral
34.
go back to reference Hartmann J, Braulke F, Sinzig U, Wulf G, Maas JH, Konietschke F, Haase D (2013) Iron overload impairs proliferation of erythroid progenitors cells (BFU-E) from patients with myelodysplastic syndromes. Leuk Res 37:327–332CrossRefPubMed Hartmann J, Braulke F, Sinzig U, Wulf G, Maas JH, Konietschke F, Haase D (2013) Iron overload impairs proliferation of erythroid progenitors cells (BFU-E) from patients with myelodysplastic syndromes. Leuk Res 37:327–332CrossRefPubMed
35.
go back to reference Taoka K, Kumano K, Nakamura F, Hosoi M, Goyama S, Imai Y, Hangaishi A, Kurokawa M (2012) The effect of iron overload and chelation on erythroid differentiation. Int J Hematol 95:149–159CrossRefPubMed Taoka K, Kumano K, Nakamura F, Hosoi M, Goyama S, Imai Y, Hangaishi A, Kurokawa M (2012) The effect of iron overload and chelation on erythroid differentiation. Int J Hematol 95:149–159CrossRefPubMed
36.
go back to reference Nicolas G, Bennoun M, Devaux I, Beaumont C, Grandchamp B, Kahn A, Vaulont S (2001) Lack of hepcidin gene expression and severe tissue iron overload in upstream stimulatory factor 2 (USF2) knockout mice. Proc Natl Acad Sci U S A 98:8780–8785CrossRefPubMedPubMedCentral Nicolas G, Bennoun M, Devaux I, Beaumont C, Grandchamp B, Kahn A, Vaulont S (2001) Lack of hepcidin gene expression and severe tissue iron overload in upstream stimulatory factor 2 (USF2) knockout mice. Proc Natl Acad Sci U S A 98:8780–8785CrossRefPubMedPubMedCentral
37.
go back to reference Pigeon C, Ilyin G, Courselaud B, Leroyer P, Turlin B, Brissot P, Loreal O (2001) A new mouse liver-specific gene, encoding a protein homologous to human antimicrobial peptide hepcidin, is overexpressed during iron overload. J Biol Chem 276:7811–7819CrossRefPubMed Pigeon C, Ilyin G, Courselaud B, Leroyer P, Turlin B, Brissot P, Loreal O (2001) A new mouse liver-specific gene, encoding a protein homologous to human antimicrobial peptide hepcidin, is overexpressed during iron overload. J Biol Chem 276:7811–7819CrossRefPubMed
39.
go back to reference Besson-Fournier C, Gineste A, Latour C, Gourbeyre O, Meynard D, Martin P, Oswald E, Coppin H, Roth MP (2017) Hepcidin upregulation by inflammation is independent of Smad1/5/8 signaling by activin B. Blood 129:533–536CrossRefPubMed Besson-Fournier C, Gineste A, Latour C, Gourbeyre O, Meynard D, Martin P, Oswald E, Coppin H, Roth MP (2017) Hepcidin upregulation by inflammation is independent of Smad1/5/8 signaling by activin B. Blood 129:533–536CrossRefPubMed
40.
go back to reference Merlevede J, Droin N, Qin T, Meldi K, Yoshida K, Morabito M, Chautard E, Auboeuf D, Fenaux P, Braun T, Itzykson R, de Botton S, Quesnel B, Commes T, Jourdan E, Vainchenker W, Bernard O, Pata-Merci N, Solier S, Gayevskiy V, Dinger ME, Cowley MJ, Selimoglu-Buet D, Meyer V, Artiguenave F, Deleuze JF, Preudhomme C, Stratton MR, Alexandrov LB, Padron E, Ogawa S, Koscielny S, Figueroa M, Solary E (2016) Mutation allele burden remains unchanged in chronic myelomonocytic leukaemia responding to hypomethylating agents. Nat Commun 7:10767CrossRefPubMedPubMedCentral Merlevede J, Droin N, Qin T, Meldi K, Yoshida K, Morabito M, Chautard E, Auboeuf D, Fenaux P, Braun T, Itzykson R, de Botton S, Quesnel B, Commes T, Jourdan E, Vainchenker W, Bernard O, Pata-Merci N, Solier S, Gayevskiy V, Dinger ME, Cowley MJ, Selimoglu-Buet D, Meyer V, Artiguenave F, Deleuze JF, Preudhomme C, Stratton MR, Alexandrov LB, Padron E, Ogawa S, Koscielny S, Figueroa M, Solary E (2016) Mutation allele burden remains unchanged in chronic myelomonocytic leukaemia responding to hypomethylating agents. Nat Commun 7:10767CrossRefPubMedPubMedCentral
Metadata
Title
Luspatercept (RAP-536) modulates oxidative stress without affecting mutation burden in myelodysplastic syndromes
Authors
Meunier Mathieu
Chloé Friedrich
Nicolas Ducrot
Johanna Zannoni
Tondeur Sylvie
Nelly Jerraya
Sophie Rousseaux
Florent Chuffart
Olivier Kosmider
Zoubida Karim
Sophie Park
Publication date
05-10-2022
Publisher
Springer Berlin Heidelberg
Published in
Annals of Hematology / Issue 12/2022
Print ISSN: 0939-5555
Electronic ISSN: 1432-0584
DOI
https://doi.org/10.1007/s00277-022-04993-7

Other articles of this Issue 12/2022

Annals of Hematology 12/2022 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine