92 results on '"Langemeijer S"'
Search Results
2. Genetic diversity within leukemia-associated immunophenotype-defined subclones in AML
- Author
-
Tiso, F., Koorenhof-Scheele, T. N., Huys, E., Martens, J. H. A., de Graaf, A. O., van der Reijden, B. A., Langemeijer, S. M. C., Preijers, F. W. M. B., Kroeze, L. I., and Jansen, J. H.
- Published
- 2022
- Full Text
- View/download PDF
3. Determinants of lenalidomide response with or without erythropoiesis-stimulating agents in myelodysplastic syndromes:the HOVON89 trial
- Author
-
van de Loosdrecht, A. A., Cremers, E. M.P., Alhan, C., Duetz, C., in ’t Hout, F. E.M., Visser-Wisselaar, H. A., Chitu, D. A., Verbrugge, A., Cunha, S. M., Ossenkoppele, G. J., Janssen, J. J.W.M., Klein, S. K., Vellenga, E., Huls, G. A., Muus, P., Langemeijer, S. M.C., de Greef, G. E., te Boekhorst, P. A.W., Raaijmakers, M. H.G., van Marwijk Kooy, M., Legdeur, M. C., Wegman, J. J., Deenik, W., de Weerdt, O., van Maanen-Lamme, T. M., Jobse, P., van Kampen, R. J.W., Beeker, A., Wijermans, P. W., Biemond, B. J., Tanis, B. C., van Esser, J. W.J., Schaar, C. G., Noordzij-Nooteboom, H. S., Jacobs, E. M.G., de Graaf, A. O., Jongen-Lavrencic, M., Stevens-Kroef, M. J.P.L., Westers, T. M., Jansen, J. H., van de Loosdrecht, A. A., Cremers, E. M.P., Alhan, C., Duetz, C., in ’t Hout, F. E.M., Visser-Wisselaar, H. A., Chitu, D. A., Verbrugge, A., Cunha, S. M., Ossenkoppele, G. J., Janssen, J. J.W.M., Klein, S. K., Vellenga, E., Huls, G. A., Muus, P., Langemeijer, S. M.C., de Greef, G. E., te Boekhorst, P. A.W., Raaijmakers, M. H.G., van Marwijk Kooy, M., Legdeur, M. C., Wegman, J. J., Deenik, W., de Weerdt, O., van Maanen-Lamme, T. M., Jobse, P., van Kampen, R. J.W., Beeker, A., Wijermans, P. W., Biemond, B. J., Tanis, B. C., van Esser, J. W.J., Schaar, C. G., Noordzij-Nooteboom, H. S., Jacobs, E. M.G., de Graaf, A. O., Jongen-Lavrencic, M., Stevens-Kroef, M. J.P.L., Westers, T. M., and Jansen, J. H.
- Abstract
A randomized phase-II study was performed in low/int-1 risk MDS (IPSS) to study efficacy and safety of lenalidomide without (arm A) or with (arm B) ESA/G-CSF. In arm B, patients without erythroid response (HI-E) after 4 cycles received ESA; G-CSF was added if no HI-E was obtained by cycle 9. HI-E served as primary endpoint. Flow cytometry and next-generation sequencing were performed to identify predictors of response. The final evaluation comprised 184 patients; 84% non-del(5q), 16% isolated del(5q); median follow-up: 70.7 months. In arm A and B, 39 and 41% of patients achieved HI-E; median time-to-HI-E: 3.2 months for both arms, median duration of-HI-E: 9.8 months. HI-E was significantly lower in non-del(5q) vs. del(5q): 32% vs. 80%. The same accounted for transfusion independency-at-week 24 (16% vs. 67%), but similar in both arms. Apart from presence of del(5q), high percentages of bone marrow lymphocytes and progenitor B-cells, a low number of mutations, absence of ring sideroblasts, and SF3B1 mutations predicted HI-E. In conclusion, lenalidomide induced HI-E in patients with non-del(5q) and del(5q) MDS without additional effect of ESA/G-CSF. The identified predictors of response may guide application of lenalidomide in lower-risk MDS in the era of precision medicine. (EudraCT 2008-002195-10).
- Published
- 2024
4. Toxic iron species in lower-risk myelodysplastic syndrome patients: course of disease and effects on outcome
- Author
-
Hoeks, M. Bagguley, T. van Marrewijk, C. Smith, A. Bowen, D. Culligan, D. Kolade, S. Symeonidis, A. Garelius, H. Spanoudakis, M. Langemeijer, S. Roelofs, R. Wiegerinck, E. Tatic, A. Killick, S. Panagiotidis, P. Stanca, O. Hellström-Lindberg, E. Cermak, J. van der Klauw, M. Wouters, H. van Kraaij, M. Blijlevens, N. Swinkels, D.W. de Witte, T. Stauder, R. Walder, A. Pfeilstöcker, M. Schoenmetzler-Makrai, A. Burgstaller, S. Thaler, J. Mandac Rogulj, I. Krejci, M. Voglova, J. Rohon, P. Jonasova, A. Cermak, J. Mikulenkova, D. Hochova, I. Jensen, P.D. Holm, M.S. Kjeldsen, L. Dufva, I.H. Vestergaard, H. Re, D. Slama, B. Fenaux, P. Choufi, B. Cheze, S. Klepping, D. Salles, B. de Renzis, B. Willems, L. De Prost, D. Gutnecht, J. Courby, S. Siguret, V. Tertian, G. Pascal, L. Chaury, M. Wattel, E. Guerci, A. Legros, L. Itzykson, R. Ades, L. Isnard, F. Sanhes, L. Benramdane, R. Stamatoullas, A. Amé, S. Beyne-Rauzy, O. Gyan, E. Platzbecker, U. Badrakan, C. Germing, U. Lübbert, M. Schlenk, R. Kotsianidis, I. Tsatalas, C. Pappa, V. Galanopoulos, A. Michali, E. Panagiotidis, P. Viniou, N. Katsigiannis, A. Roussou, P. Terpos, E. Kostourou, A. Kartasis, Z. Pouli, A. Palla, K. Briasoulis, V. Hatzimichael, E. Vassilopoulos, G. Symeonidis, A. Kourakli, A. Zikos, P. Anagnostopoulos, A. Kotsopoulou, M. Megalakaki, K. Protopapa, M. Vlachaki, E. Konstantinidou, P. Stemer, G. Nemetz, A. Gotwin, U. Cohen, O. Koren, M. Levy, E. Greenbaum, U. Gino-Moor, S. Price, M. Ofran, Y. Winder, A. Goldshmidt, N. Elias, S. Sabag, R. Hellman, I. Ellis, M. Braester, A. Rosenbaum, H. Berdichevsky, S. Itzhaki, G. Wolaj, O. Yeganeh, S. Katz, O. Filanovsky, K. Dali, N. Mittelman, M. Malcovati, L. Fianchi, L. vd Loosdrecht, A. Matthijssen, V. Herbers, A. Pruijt, H. Aboosy, N. de Vries, F. Velders, G. Jacobs, E. Langemeijer, S. MacKenzie, M. Lensen, C. Kuijper, P. Madry, K. Camara, M. Almeida, A. Vulkan, G. Stanca Ciocan, O. Tatic, A. Savic, A. Pedro, C. Xicoy, B. Leiva, P. Munoz, J. Betes, V. Benavente, C. Lozano and Hoeks, M. Bagguley, T. van Marrewijk, C. Smith, A. Bowen, D. Culligan, D. Kolade, S. Symeonidis, A. Garelius, H. Spanoudakis, M. Langemeijer, S. Roelofs, R. Wiegerinck, E. Tatic, A. Killick, S. Panagiotidis, P. Stanca, O. Hellström-Lindberg, E. Cermak, J. van der Klauw, M. Wouters, H. van Kraaij, M. Blijlevens, N. Swinkels, D.W. de Witte, T. Stauder, R. Walder, A. Pfeilstöcker, M. Schoenmetzler-Makrai, A. Burgstaller, S. Thaler, J. Mandac Rogulj, I. Krejci, M. Voglova, J. Rohon, P. Jonasova, A. Cermak, J. Mikulenkova, D. Hochova, I. Jensen, P.D. Holm, M.S. Kjeldsen, L. Dufva, I.H. Vestergaard, H. Re, D. Slama, B. Fenaux, P. Choufi, B. Cheze, S. Klepping, D. Salles, B. de Renzis, B. Willems, L. De Prost, D. Gutnecht, J. Courby, S. Siguret, V. Tertian, G. Pascal, L. Chaury, M. Wattel, E. Guerci, A. Legros, L. Itzykson, R. Ades, L. Isnard, F. Sanhes, L. Benramdane, R. Stamatoullas, A. Amé, S. Beyne-Rauzy, O. Gyan, E. Platzbecker, U. Badrakan, C. Germing, U. Lübbert, M. Schlenk, R. Kotsianidis, I. Tsatalas, C. Pappa, V. Galanopoulos, A. Michali, E. Panagiotidis, P. Viniou, N. Katsigiannis, A. Roussou, P. Terpos, E. Kostourou, A. Kartasis, Z. Pouli, A. Palla, K. Briasoulis, V. Hatzimichael, E. Vassilopoulos, G. Symeonidis, A. Kourakli, A. Zikos, P. Anagnostopoulos, A. Kotsopoulou, M. Megalakaki, K. Protopapa, M. Vlachaki, E. Konstantinidou, P. Stemer, G. Nemetz, A. Gotwin, U. Cohen, O. Koren, M. Levy, E. Greenbaum, U. Gino-Moor, S. Price, M. Ofran, Y. Winder, A. Goldshmidt, N. Elias, S. Sabag, R. Hellman, I. Ellis, M. Braester, A. Rosenbaum, H. Berdichevsky, S. Itzhaki, G. Wolaj, O. Yeganeh, S. Katz, O. Filanovsky, K. Dali, N. Mittelman, M. Malcovati, L. Fianchi, L. vd Loosdrecht, A. Matthijssen, V. Herbers, A. Pruijt, H. Aboosy, N. de Vries, F. Velders, G. Jacobs, E. Langemeijer, S. MacKenzie, M. Lensen, C. Kuijper, P. Madry, K. Camara, M. Almeida, A. Vulkan, G. Stanca Ciocan, O. Tatic, A. Savic, A. Pedro, C. Xicoy, B. Leiva, P. Munoz, J. Betes, V. Benavente, C. Lozano
- Published
- 2021
5. S168: ERYTHROPOIETIN STIMULATION AGENTS SIGNIFICANTLY IMPROVES OUTCOME IN LOWER RISK MDS.
- Author
-
Garelius, H., primary, Smith, A., additional, Bagguley, T., additional, Taylor, A., additional, Fenaux, P., additional, Bowen, D., additional, Symeonidis, A., additional, Mittelmann, M., additional, Stauder, R., additional, Čermák, J., additional, Sanz, G., additional, Langemeijer, S., additional, Malcovati, L., additional, Germing, U., additional, Itzykson, R., additional, Guerci-Bresler, A., additional, Culligan, D., additional, Kotsianidis, I., additional, Koinig Mag, K., additional, van Marrewijk, C., additional, Crouch, S., additional, de Witte, T., additional, and Hellström-Lindberg, E., additional
- Published
- 2022
- Full Text
- View/download PDF
6. P471: ERYTHROID/MEGAKARYOCYTIC DIFFERENTIATION BIAS IN BONE MARROW OF AML/MDS PATIENTS AFTER DECITABINE TREATMENT
- Author
-
Tiso, F., primary, de Graaf, A. O., additional, Kroeze, L. I., additional, van der Reijden, B. A., additional, Langemeijer, S. M. C., additional, Hebeda, K. M., additional, and Jansen, J. H., additional
- Published
- 2022
- Full Text
- View/download PDF
7. Toxic iron species in lower-risk myelodysplastic syndrome patients: course of disease and effects on outcome
- Author
-
Hoeks, M. Bagguley, T. van Marrewijk, C. Smith, A. Bowen, D. Culligan, D. Kolade, S. Symeonidis, A. Garelius, H. Spanoudakis, M. Langemeijer, S. Roelofs, R. Wiegerinck, E. Tatic, A. Killick, S. Panagiotidis, P. Stanca, O. Hellström-Lindberg, E. Cermak, J. van der Klauw, M. Wouters, H. van Kraaij, M. Blijlevens, N. Swinkels, D.W. de Witte, T. Stauder, R. Walder, A. Pfeilstöcker, M. Schoenmetzler-Makrai, A. Burgstaller, S. Thaler, J. Mandac Rogulj, I. Krejci, M. Voglova, J. Rohon, P. Jonasova, A. Cermak, J. Mikulenkova, D. Hochova, I. Jensen, P.D. Holm, M.S. Kjeldsen, L. Dufva, I.H. Vestergaard, H. Re, D. Slama, B. Fenaux, P. Choufi, B. Cheze, S. Klepping, D. Salles, B. de Renzis, B. Willems, L. De Prost, D. Gutnecht, J. Courby, S. Siguret, V. Tertian, G. Pascal, L. Chaury, M. Wattel, E. Guerci, A. Legros, L. Itzykson, R. Ades, L. Isnard, F. Sanhes, L. Benramdane, R. Stamatoullas, A. Amé, S. Beyne-Rauzy, O. Gyan, E. Platzbecker, U. Badrakan, C. Germing, U. Lübbert, M. Schlenk, R. Kotsianidis, I. Tsatalas, C. Pappa, V. Galanopoulos, A. Michali, E. Panagiotidis, P. Viniou, N. Katsigiannis, A. Roussou, P. Terpos, E. Kostourou, A. Kartasis, Z. Pouli, A. Palla, K. Briasoulis, V. Hatzimichael, E. Vassilopoulos, G. Symeonidis, A. Kourakli, A. Zikos, P. Anagnostopoulos, A. Kotsopoulou, M. Megalakaki, K. Protopapa, M. Vlachaki, E. Konstantinidou, P. Stemer, G. Nemetz, A. Gotwin, U. Cohen, O. Koren, M. Levy, E. Greenbaum, U. Gino-Moor, S. Price, M. Ofran, Y. Winder, A. Goldshmidt, N. Elias, S. Sabag, R. Hellman, I. Ellis, M. Braester, A. Rosenbaum, H. Berdichevsky, S. Itzhaki, G. Wolaj, O. Yeganeh, S. Katz, O. Filanovsky, K. Dali, N. Mittelman, M. Malcovati, L. Fianchi, L. vd Loosdrecht, A. Matthijssen, V. Herbers, A. Pruijt, H. Aboosy, N. de Vries, F. Velders, G. Jacobs, E. Langemeijer, S. MacKenzie, M. Lensen, C. Kuijper, P. Madry, K. Camara, M. Almeida, A. Vulkan, G. Stanca Ciocan, O. Tatic, A. Savic, A. Pedro, C. Xicoy, B. Leiva, P. Munoz, J. Betes, V. Benavente, C. Lozano, M. Martinez, M. Iniesta, P. Bernal, T. Diez Campelo, M. Tormo, D. Andreu Lapiedra, R. Sanz, G. Hesse Sundin, E. Garelius, H. Karlsson, C. Antunovic, P. Jönsson, A. Brandefors, L. Nilsson, L. Kozlowski, P. Hellstrom-Lindberg, E. Grövdal, M. Larsson, K. Wallvik, J. Lorenz, F. Ejerblad, E. Culligan, D. Craddock, C. Kolade, S. Cahalin, P. Killick, S. Ackroyd, S. Wong, C. Warren, A. Drummond, M. Hall, C. Rothwell, K. Green, S. Ali, S. Karakantza, M. Dennis, M. Jones, G. Parker, J. Bowen, A. Radia, R. Das-Gupta, E. Vyas, P. Nga, E. Creagh, D. Ashcroft, J. Mills, J. Bond, L. the EUMDS Registry Participants
- Published
- 2021
8. P32 - Topic: AS04-MDS Biology and Pathogenesis/AS04h-Immune deregulation: HIGH-DIMENSIONAL PROFILING OF T AND NK CELL FREQUENCIES AND UNSUPERVISED CLUSTERING ANALYSES TO IDENTIFY IMMUNE (DYS) FUNCTIONAL PHENOTYPES IN MDS PATIENTS
- Author
-
Van Ens, D., Brummelman, J., Van Eck Van Der Sluijs, J., Woestenenk, R., Heister, D., Koorenhof-Scheele, T., Langemeijer, S., Jansen, J., Dolstra, H., and Hobo, W.
- Published
- 2021
- Full Text
- View/download PDF
9. O08 - Topic: AS06-Prognosis/AS06a-Prognostic factors of outcome and risk assessment: LYMPHOPENIA IS HIGHLY PREVALENT IN MDS AND PROVIDES ADDITIONAL PROGNOSTIC INFORMATION FOR IPSS-R VERY-LOW AND LOW-RISK PATIENTS. AN ANALYSIS FROM THE EU-MDS REGISTRY
- Author
-
Silzle, T., Taylor, A., De Witte, T., Malcovati, L., Fenaux, P., Bowen, D., Symeonidis, A., Mittelman, M., Stauder, R., Cermak, J., Sanz, G., Hellström-Lindberg, E., Langemeijer, S., Holm, M. Skov, Madry, K., Tatic, A., Almeida, A. Medina, Savic, A., Rogulj, I. Mandac, Germing, U., and Smith, A.
- Published
- 2021
- Full Text
- View/download PDF
10. Toxic iron species in lower-risk myelodysplastic syndrome patients : course of disease and effects on outcome
- Author
-
Hoeks, Marlijn, Bagguley, Tim, van Marrewijk, Corine, Smith, Alex, Bowen, David, Culligan, Dominic, Kolade, Seye, Symeonidis, Argiris, Garelius, Hege, Spanoudakis, Michail, Langemeijer, Saskia, Roelofs, Rian, Wiegerinck, Erwin, Tatic, Aurelia, Killick, Sally, Panagiotidis, Panagiotis, Stanca, Oana, Hellström-Lindberg, Eva, Cermak, Jaroslav, van der Klauw, Melanie, Wouters, Hanneke, van Kraaij, Marian, Blijlevens, Nicole, Swinkels, Dorine W., de Witte, Theo, Stauder, R., Walder, A., Pfeilstöcker, M., Schoenmetzler-Makrai, A., Burgstaller, S., Thaler, J., Mandac Rogulj, I., Krejci, M., Voglova, J., Rohon, P., Jonasova, A., Cermak, J., Mikulenkova, D., Hochova, I., Jensen, P. D., Holm, M. S., Kjeldsen, L., Dufva, I. H., Vestergaard, H., Re, D., Slama, B., Fenaux, P., Choufi, B., Cheze, S., Klepping, D., Salles, B., de Renzis, B., Willems, L., De Prost, D., Gutnecht, J., Courby, S., Siguret, V., Tertian, G., Pascal, L., Chaury, M., Wattel, E., Guerci, A., Legros, L., Itzykson, R., Ades, L., Isnard, F., Sanhes, L., Benramdane, R., Stamatoullas, A., Amé, S., Beyne-Rauzy, O., Gyan, E., Platzbecker, U., Badrakan, C., Germing, U., Lübbert, M., Schlenk, R., Kotsianidis, I., Tsatalas, C., Pappa, V., Galanopoulos, A., Michali, E., Panagiotidis, P., Viniou, N., Katsigiannis, A., Roussou, P., Terpos, E., Kostourou, A., Kartasis, Z., Pouli, A., Palla, K., Briasoulis, V., Hatzimichael, E., Vassilopoulos, G., Symeonidis, A., Kourakli, A., Zikos, P., Anagnostopoulos, A., Kotsopoulou, M., Megalakaki, K., Protopapa, M., Vlachaki, E., Konstantinidou, P., Stemer, G., Nemetz, A., Gotwin, U., Cohen, O., Koren, M., Levy, E., Greenbaum, U., Gino-Moor, S., Price, M., Ofran, Y., Winder, A., Goldshmidt, N., Elias, S., Sabag, R., Hellman, I., Ellis, M., Braester, A., Rosenbaum, H., Berdichevsky, S., Itzhaki, G., Wolaj, O., Yeganeh, S., Katz, O., Filanovsky, K., Dali, N., Mittelman, M., Malcovati, L., Fianchi, L., vd Loosdrecht, A., Matthijssen, V., Herbers, A., Pruijt, H., Aboosy, N., de Vries, F., Velders, G., Jacobs, E., Langemeijer, S., MacKenzie, M., Lensen, C., Kuijper, P., Madry, K., Camara, M., Almeida, A., Vulkan, G., Stanca Ciocan, O., Tatic, A., Savic, A., Pedro, C., Xicoy, B., Leiva, P., Munoz, J., Betes, V., Benavente, C., Lozano, M., Martinez, M., Iniesta, P., Bernal, T., Diez Campelo, M., Tormo, D., Andreu Lapiedra, R., Sanz, G., Hesse Sundin, E., Garelius, H., Karlsson, C., Antunovic, P., Jönsson, A., Brandefors, L., Nilsson, L., Kozlowski, P., Hellstrom-Lindberg, E., Grövdal, M., Larsson, K., Wallvik, J., Lorenz, F., Ejerblad, E., Culligan, D., Craddock, C., Kolade, S., Cahalin, P., Killick, S., Ackroyd, S., Wong, C., Warren, A., Drummond, M., Hall, C., Rothwell, K., Green, S., Ali, S., Bowen, D., Karakantza, M., Dennis, M., Jones, G., Parker, J., Bowen, A., Radia, R., Das-Gupta, E., Vyas, P., Nga, E., Creagh, D., Ashcroft, J., Mills, J., Bond, L., Life Course Epidemiology (LCE), and VU University medical center
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Iron Overload ,Iron ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,Vascular damage Radboud Institute for Health Sciences [Radboudumc 16] ,Ferroportin ,Lower risk ,Gastroenterology ,Article ,Cancer development and immune defence Radboud Institute for Health Sciences [Radboudumc 2] ,03 medical and health sciences ,0302 clinical medicine ,Hepcidin ,Internal medicine ,Humans ,Medicine ,Blood Transfusion ,Prospective Studies ,Aged ,Soluble transferrin receptor ,biology ,business.industry ,Transferrin saturation ,Hematology ,Middle Aged ,Erythroferrone ,Prognosis ,3. Good health ,Survival Rate ,Ferritin ,Renal disorders Radboud Institute for Molecular Life Sciences [Radboudumc 11] ,030104 developmental biology ,Oncology ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,biology.protein ,Erythropoiesis ,Female ,business ,Myelodysplastic syndrome ,Follow-Up Studies ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] - Abstract
Red blood cell transfusions (RBCT) remain the cornerstone of supportive care in lower-risk myelodysplastic syndrome (LRMDS) [1]. Transfusion dependency in LRMDS patients is associated with inferior outcomes, mainly attributed to severe bone marrow failure [2]. However, iron toxicity, due to frequent RBCT or ineffective erythropoiesis, may be an additional negative prognostic factor [3,4,5,6]. Recently, much progress has been made in unraveling the iron metabolism. The peptide hormone hepcidin is the key regulator by inhibiting iron uptake through degradation of ferroportin, a cellular iron exporter [7]. Erythroferrone and GDF15, produced by erythroblasts, inhibit hepcidin production, which leads to increased uptake and cellular release of iron for the purpose of erythropoiesis [8]. The pathophysiology of iron metabolism in MDS is still not completely understood. Exceedingly high reactive oxygen species (ROS) levels are associated with iron toxicity, disease development, and progression in MDS patients [9,10,11,12]. Malondialdehyde (MDA), resulting from lipid peroxidation of polyunsaturated fatty acids, is a biomarker of oxidative stress [10, 12]. Currently, little is known about the prognostic impact of ROS in MDS patients. The aim of this study is twofold: (1) describe iron and oxidative stress parameters over time in LRMDS patients and (2) to assess their effect on overall and progression-free survival. The EUMDS registry prospectively collects observational data on newly diagnosed LRMDS patients from 148 centers in 16 countries in Europe and Israel as of January 2008. All patients provided informed consent. Clinical data were collected at baseline and at each six-monthly follow-up visit. Serum samples were collected prospectively at each visit from 256 patients included in six participating countries. Conventional iron parameters were measured with routine assays. We additionally analyzed hepcidin, growth differentiation factor 15 (GDF15), soluble transferrin receptor (sTfR), non-transferrin bound iron (NTBI), labile plasma iron (LPI), and MDA. Subjects were prospectively followed until death, loss to follow-up, or withdrawal of consent. All iron parameters were measured centrally at the department of Laboratory Medicine of the Radboudumc, Nijmegen, The Netherlands. Serum samples were collected just prior to transfusion in transfusion-dependent patients and stored at −80 °C. Details on the assays and reference ranges of hepcidin, GDF15, sTfR, NTBI, LPI, and MDA are provided in the supplement. The Spearman rank test was used to evaluate correlations between iron parameters. We stratified the results by transfusion dependency per visit and the presence of ring sideroblasts. When evaluating temporal changes in iron parameters, with linear quantile mixed models, we excluded patients from the timepoint they received iron chelation therapy. Overall survival (OS) was defined as the time from MDS diagnosis to death or, in case of progression-free survival, to date of progression or death; patients still alive at the end of follow-up were censored. Time-dependent Kaplan–Meier curves and cox proportional hazards models were used. In total, 256 consecutive patients, were included in this study. Over five six-monthly visits, 1040 samples were collected. Table 1 describes the patient characteristics. Most patients without ring sideroblasts were transfusion-independent at diagnosis (nonRS-TI; 55.9%), 18.8% with ring sideroblasts were transfusion-independent (RS-TI), 18.4% without ring sideroblasts were transfusion-dependent (nonRS-TD), and 7% with ring sideroblasts were transfusion-dependent patients (RS-TD). The median follow-up time was 6.6 years (95% CI 5.9–7.0). LPI was positively correlated with transferrin saturation (TSAT) (r = 0.15, p < 0.001, Fig. S1). LPI values increased exponentially at TSAT values above 80%. This effect was most pronounced in the transfusion-dependent groups, but also observed in the RS-TI group. MDA was weakly correlated with NTBI (r = 0.09, p = 0.069) and negatively correlated with hemoglobin level (r = −0.1, p = 0.033). GDF15 and hepcidin were negatively correlated in the RS-TI and nonRS-TD group and significantly negatively correlated in the RS-TD group (r = −0.34, p = 0.007, Fig. S2). Serum ferritin levels were elevated in all subgroups with a mean value of 858 µg/L at visit 5. The highest serum ferritin levels were observed in the RS-TD group (mean value at visit 5: 2092 µg/L, Table S1). Serum ferritin increased significantly per visit in the RS-TD group (beta 454.46 µg/L; 95% CI 334.65–574.27), but not in the other groups (Table S2). All subgroups, except for the nonRS-TI, had elevated TSAT levels. TSAT levels were most markedly increased in the RS-TD group with a mean TSAT of 88% at visit 5 (Table S1). In both transfusion-dependent groups the median increase per visit was significant (Table S2). LPI was elevated in the RS-TD group exclusively with a mean value of 0.59 µmol/L at visit 5 (Table S1). NTBI was elevated in all subgroups, with the highest values in the RS-TD group (Table S1). The increase in median NTBI level was significant in both transfusion-dependent groups (Table S2). Hepcidin levels were markedly elevated in the nonRS-TD group. Interestingly, hepcidin levels were lower in the RS-TD group, probably reflecting ineffective erythropoiesis, likewise supported by lower hepcidin/ferritin ratios in RS groups (Table S1). Median hepcidin levels increased over time in the transfusion-dependent subgroups only (Table S2). GDF15 levels, analyzed in the light of its potential role in hepcidin suppression, were increased in all subgroups (Table S1). The RS subgroups had higher GDF15 levels compared to the nonRS groups, reflecting increased erythropoiesis. Mean sTfR levels were within the reference range in all subgroups except for the RS-TI group, which showed elevated levels, reflecting...
- Published
- 2021
- Full Text
- View/download PDF
11. Topic: AS06-Prognosis/AS06a-Prognostic factors of outcome and risk assessment
- Author
-
Silzle, T., primary, Taylor, A., additional, De Witte, T., additional, Malcovati, L., additional, Fenaux, P., additional, Bowen, D., additional, Symeonidis, A., additional, Mittelman, M., additional, Stauder, R., additional, Cermak, J., additional, Sanz, G., additional, Hellström-Lindberg, E., additional, Langemeijer, S., additional, Holm, M. Skov, additional, Madry, K., additional, Tatic, A., additional, Almeida, A. Medina, additional, Savic, A., additional, Rogulj, I. Mandac, additional, Germing, U., additional, and Smith, A., additional
- Published
- 2021
- Full Text
- View/download PDF
12. TET2 mutations in childhood leukemia
- Author
-
Langemeijer, S M C, Jansen, J H, Hooijer, J, van Hoogen, P, Stevens-Linders, E, Massop, M, Waanders, E, van Reijmersdal, S V, Stevens-Kroef, M J P L, Zwaan, C M, van den Heuvel-Eibrink, M M, Sonneveld, E, Hoogerbrugge, P M, Geurts van Kessel, A, and Kuiper, R P
- Published
- 2011
- Full Text
- View/download PDF
13. New cases of adult-onset Sandhoff disease with a cerebellar or lower motor neuron phenotype
- Author
-
Delnooz, C C, Lefeber, D J, Langemeijer, S M, Hoffjan, S, Dekomien, G, Zwarts, M J, Van Engelen, B G, Wevers, R A, Schelhaas, H J, and van de Warrenburg, B P
- Published
- 2010
- Full Text
- View/download PDF
14. Impact of red blood cell transfusion dose density on progression-free survival in lower-risk myelodysplastic syndromes patients
- Author
-
de Swart L, Crouch S, Hoeks M, Smith A, Langemeijer S, Fenaux P, Symeonidis A, Cermák J, Hellström-Lindberg E, Stauder R, Sanz G, Mittelman M, Holm MS, Malcovati L, Madry K, Germing U, Tatic A, Savic A, Almeida AM, Gredelj-Šimec N, Guerci-Bresler A, Beyne-Rauzy O, Culligan D, Kotsianidis I, Itzykson R, van Marrewijk C, Blijlevens N, Bowen D, de Witte T, and EUMDS Registry Participants
- Subjects
Myelodysplastic Syndromes, lower-risk, progression-free survival, red blood cell transfusions, transfusion dose density - Abstract
Progression-free survival of lower-risk myelodysplastic syndromes patients treated with red blood cell transfusions is usually reduced, but it is unclear whether transfusion dose density is an independent prognostic factor. The European MDS Registry collects prospective data at 6-monthly intervals of newly diagnosed lower-risk myelodysplastic syndromes patients from 16 European countries and Israel. Data on the transfusion dose density - the cumulative dose received at the end of each interval divided by the time since the beginning of the interval in which the first transfusion was received - were analyzed using proportional hazards regression with time-varying co-variates, with death and progression to higher-risk myelodysplastic syndromes /acute myeloid leukemia as events. Of the 1267 patients included in the analyses, 317 patients died without progression, in 162 patients the disease had progressed. Progression-free survival was significantly associated with age, EQ-5D index, baseline WHO classification, bone marrow blast count, cytogenetic risk category, number of cytopenias, and country. Transfusion dose density was inversely associated with progression-free survival (p1x10-4): dose density had an increasing effect on hazard until a dose density of 3 units/16 weeks. The transfusion dose density effect continued to increase beyond 8 units/16 weeks after correction for the impact of treatment with erythropoietin agents, lenalidomide and/or iron chelators. Conclusion: the negative effect of transfusion treatment on progression-free survival already occurs at transfusion densities below 3 units/16 weeks. This indicates that transfusion dependency, even at relatively low dose densities, may be considered as an indicator of inferior progression-free survival. This trial was registered at www.clinicaltrials.gov as #NCT00600860.
- Published
- 2020
15. Successful treatment of a PNH patient non-responsive to eculizumab with the novel complement C5 inhibitor coversin (nomacopan)
- Author
-
Schols, S.E.M., Nunn, M.A., Mackie, I., Weston-Davies, W., Nishimura, J.I., Kanakura, Y., Blijlevens, N.M., Muus, P., Langemeijer, S., Schols, S.E.M., Nunn, M.A., Mackie, I., Weston-Davies, W., Nishimura, J.I., Kanakura, Y., Blijlevens, N.M., Muus, P., and Langemeijer, S.
- Abstract
Contains fulltext : 218318.pdf (Publisher’s version ) (Closed access)
- Published
- 2020
16. S838 TRANSFUSION DEPENDENCY IS ASSOCIATED WITH PRESENCE OF TOXIC IRON SPECIES AND INFERIOR SURVIVAL IN PATIENTS WITH LOWER-RISK MYELODYSPLASTIC SYNDROMES
- Author
-
Hoeks, M., primary, Bagguley, T., additional, Roelofs, R., additional, de Swart, L., additional, Bowen, D., additional, Symeonidis, A., additional, van Marrewijk, C., additional, Hellstrom-Lindberg, E., additional, Tatic, A., additional, Langemeijer, S., additional, Culligan, D., additional, Macheta, M., additional, Garelius, H., additional, Spanoudakis, M., additional, Killick, S., additional, Panagiotidis, P., additional, Stanca Ciocan, O., additional, Mills, J., additional, Pottinger, B., additional, Ackroyd, S., additional, Hall, C., additional, Droste, J., additional, Smith, A., additional, Swinkels, D., additional, and de Witte, T., additional
- Published
- 2019
- Full Text
- View/download PDF
17. PS1121 ABSENCE OF GPI-NEGATIVE CELLS AT DIAGNOSIS AND LOW NEUTROPHIL COUNT 2 MONTHS AFTER ATGAM BASED TREATMENT ARE ASSOCIATED WITH LOWER 6-MONTH RESPONSE RATE IN ADULTS WITH (VERY) SEVERE APLASTIC ANEMIA
- Author
-
Tjon, J., primary, Bogers, L., additional, Wreede, L. de, additional, Groot, M. de, additional, Langemeijer, S., additional, Koene, H., additional, Meijer, E., additional, Nur, E., additional, Raaijmakers, M., additional, Raymakers, R., additional, Snijders, T., additional, and Halkes, S., additional
- Published
- 2019
- Full Text
- View/download PDF
18. Labile plasma iron levels predict survival in patients with lower-risk myelodysplastic syndromes
- Author
-
de Swart, L. Reiniers, C. Bagguley, T. van Marrewijk, C. Bowen, D. Hellström-Lindberg, E. Tatic, A. Symeonidis, A. Huls, G. Cermak, J. van de Loosdrecht, A.A. Garelius, H. Culligan, D. Macheta, M. Spanoudakis, M. Panagiotidis, P. Krejci, M. Blijlevens, N. Langemeijer, S. Droste, J. Swinkels, D.W. Smith, A. de Witte, T. EUMDS Steering Committee
- Abstract
Red blood cell transfusions remain one of the cornerstones in supportive care of lower-risk patients with myelodysplastic syndromes. We hypothesized that patients develop oxidant-mediated tissue injury through the formation of toxic iron species, caused either by red blood cell transfusions or by ineffective erythropoiesis. We analyzed serum samples from 100 lower-risk patients with myelodysplastic syndromes at six-month intervals for transferrin saturation, hepcidin-25, growth differentiation factor 15, soluble transferrin receptor, non-transferrin bound iron and labile plasma iron in order to evaluate temporal changes in iron metabolism and the presence of potentially toxic iron species and their impact on survival. Hepcidin levels were low in 34 patients with ringed sideroblasts compared to 66 patients without. Increases of hepcidin and non-transferrin bound iron levels were visible early in follow-up of all transfusion-dependent patient groups. Hepcidin levels significantly decreased over time in transfusion-independent patients with ringed sideroblasts. Increased soluble transferrin receptor levels in transfusion-independent patients with ringed sideroblasts confirmed the presence of ineffective erythropoiesis and suppression of hepcidin production in these patients. Detectable labile plasma iron levels in combination with high transferrin saturation levels occurred almost exclusively in patients with ringed sideroblasts and all transfusiondependent patient groups. Detectable labile plasma iron levels in transfusion-dependent patients without ringed sideroblasts were associated with decreased survival. In conclusion, toxic iron species occurred in all transfusion-dependent patients and in transfusion-independent patients with ringed sideroblasts. Labile plasma iron appeared to be a clinically relevant measure for potential iron toxicity and a prognostic factor for survival in transfusion-dependent patients. © 2018 Ferrata Storti Foundation.
- Published
- 2018
19. TREATMENT WITH HORSE-DERIVED ANTI-THYMOCYTE GLOBULIN LEADS TO ENDURING HEMATOLOGICAL RESPONSES AND A 1.5-YEAR SURVIVAL PROBABILITY OF 87% IN ADULT ACQUIRED APLASTIC ANEMIA PATIENTS IN THE NETHERLANDS
- Author
-
Tjon, J., Wreede, L. de, Falkenburg, F., Groot, M. de, Koene, H., Langemeijer, S., Meijer, E., Raaijmakers, M., Raymakers, R., Snijders, T., Zeerleder, S., and Halkes, S.
- Published
- 2017
20. P052 Acquired mutations in TET2 are common in myelodysplastic syndromes
- Author
-
Langemeijer, S., Kuiper, R., Berends, M., Knops, R., Aslanyan, M., Massop, M., van Hoogen, P., van Kessel, A. Geurts, Raymakers, R., Verburgh, E., Hagemeijer, A., Vandenberghe, P., de Witte, T., Van der Reijden, B., and Jansen, J.
- Published
- 2009
- Full Text
- View/download PDF
21. Impact of Red Blood Cell Transfusions on Survival in Lower-Risk MDS Patients Included in the European Leukemianet MDS (EUMDS) Registry
- Author
-
de Swart, L., primary, Crouch, S., additional, Smith, A., additional, Fenaux, P., additional, Symeonidis, A., additional, Cermak, J., additional, Hellström-Lindberg, E., additional, Sanz, G., additional, Stauder, R., additional, Malcovati, L., additional, Germing, U., additional, Langemeijer, S., additional, Skov Holm, M., additional, Mittelman, M., additional, Mądry, K., additional, Almeida, A., additional, Savic, A., additional, Itzykson, R., additional, Bowen, D., additional, and de Witte, T., additional
- Published
- 2017
- Full Text
- View/download PDF
22. Prognostic Value of Early Drop in Platelets in Lower-Risk MDS. A Sub-Study from the European Leukemianet Lower-Risk MDS (EUMDS) Registry
- Author
-
Itzykson, R., primary, Smith, A., additional, Fenaux, P., additional, Symeonidis, A., additional, Mittelman, M., additional, Stauder, R., additional, Sanz, G., additional, Čermák, J., additional, Hellström-Lindberg, E., additional, Malcovati, L., additional, Langemeijer, S., additional, Germing, U., additional, Mądry, K., additional, Holm, M.S., additional, Tatic, A., additional, Almeida, A.M., additional, Savic, A., additional, Gredelj Šimec, N., additional, Bowen, D., additional, and de Witte, T., additional
- Published
- 2017
- Full Text
- View/download PDF
23. Health-Related Quality of Life is Substantially Impaired in Lower-Risk MDS when Compared with Reference Populations and Significantly Affects Overall Survival
- Author
-
Stauder, R., primary, Yu, G., additional, Koinig, K., additional, Fenaux, P., additional, Symeonidis, A., additional, Sanz, G., additional, Cermak, J., additional, Mittelman, M., additional, Hellström-Lindberg, E., additional, Langemeijer, S., additional, Skov Holm, M., additional, Mądry, K., additional, Malcovati, L., additional, Tatic, A., additional, Germing, U., additional, Savic, A., additional, Efficace, F., additional, Smith, A., additional, Bowen, D., additional, and de Witte, T., additional
- Published
- 2017
- Full Text
- View/download PDF
24. 280 - Impact of Red Blood Cell Transfusions on Survival in Lower-Risk MDS Patients Included in the European Leukemianet MDS (EUMDS) Registry
- Author
-
de Swart, L., Crouch, S., Smith, A., Fenaux, P., Symeonidis, A., Cermak, J., Hellström-Lindberg, E., Sanz, G., Stauder, R., Malcovati, L., Germing, U., Langemeijer, S., Skov Holm, M., Mittelman, M., Mądry, K., Almeida, A., Savic, A., Itzykson, R., Bowen, D., and de Witte, T.
- Published
- 2017
- Full Text
- View/download PDF
25. 22 - Health-Related Quality of Life is Substantially Impaired in Lower-Risk MDS when Compared with Reference Populations and Significantly Affects Overall Survival
- Author
-
Stauder, R., Yu, G., Koinig, K., Fenaux, P., Symeonidis, A., Sanz, G., Cermak, J., Mittelman, M., Hellström-Lindberg, E., Langemeijer, S., Skov Holm, M., Mądry, K., Malcovati, L., Tatic, A., Germing, U., Savic, A., Efficace, F., Smith, A., Bowen, D., and de Witte, T.
- Published
- 2017
- Full Text
- View/download PDF
26. 19 - Prognostic Value of Early Drop in Platelets in Lower-Risk MDS. A Sub-Study from the European Leukemianet Lower-Risk MDS (EUMDS) Registry
- Author
-
Itzykson, R., Smith, A., Fenaux, P., Symeonidis, A., Mittelman, M., Stauder, R., Sanz, G., Čermák, J., Hellström-Lindberg, E., Malcovati, L., Langemeijer, S., Germing, U., Mądry, K., Holm, M.S., Tatic, A., Almeida, A.M., Savic, A., Gredelj Šimec, N., Bowen, D., and de Witte, T.
- Published
- 2017
- Full Text
- View/download PDF
27. TET2 mutations in childhood leukemia
- Author
-
Langemeijer, S M C, primary, Jansen, J H, additional, Hooijer, J, additional, van Hoogen, P, additional, Stevens-Linders, E, additional, Massop, M, additional, Waanders, E, additional, van Reijmersdal, S V, additional, Stevens-Kroef, M J P L, additional, Zwaan, C M, additional, van den Heuvel-Eibrink, M M, additional, Sonneveld, E, additional, Hoogerbrugge, P M, additional, Geurts van Kessel, A, additional, and Kuiper, R P, additional
- Published
- 2010
- Full Text
- View/download PDF
28. Incidence and Clinical Impact of TET2 Mutations in Acute Myeloid Leukemia Patients Treated within the EORTC/GIMEMA AML-12/06991 AML Trial.
- Author
-
Aslanyan, M. G, primary, Langemeijer, S. M.C., additional, Cilloni, D., additional, Saglio, G., additional, Marie, JP, additional, Tang, R., additional, Labar, B., additional, Zadro, R., additional, Batinic, D., additional, Amadori, S., additional, Lo Coco, F., additional, Scheele, T., additional, Kroeze, L., additional, Massop, M., additional, van Hoogen, P., additional, Stevens, E., additional, Muus, P., additional, Suciu, S., additional, Baila, L., additional, Marijt, E. W.A., additional, Willemze, R., additional, de Witte, T., additional, van der Reijden, B., additional, and Jansen, J. H., additional
- Published
- 2009
- Full Text
- View/download PDF
29. C020 Identification of novel genetic lesions in MDS using high density SNP-arrays
- Author
-
Langemeijer, S., primary, Kuiper, R., additional, vd Reijden, B.A., additional, de Witte, T., additional, and Jansen, J.H., additional
- Published
- 2007
- Full Text
- View/download PDF
30. COMPARISON OF 1690 MDS PATIENTS FROM THE EUROPEAN LEUKEMIANET REGISTRY AND REFERENCE POPULATIONS - EVIDENCE FOR A SIGNIFICANT IMPAIRMENT IN QOL IN MDS AND DEFINITION OF PREDICTORS OF DIMINISHED QOL
- Author
-
Stauder, R., Yu, G., Bagguley, T., Fenaux, P., Symeonidis, A., Sanz, G., Cermak, J., Mittelman, M., Hellstrom-Lindberg, E., Langemeijer, S., Holm, M. S., Madry, K., LUCA MALCOVATI, Tatic, A., Germing, U., Savic, A., Marrewijk, C., Guerci-Bresler, A., Luno, E., Droste, J., Koinig, K., Smith, A., Bowen, D., and Witte, T.
31. HEALTH-RELATEDQUALITYOF LIFE IS SUBSTANTIALLY IMPAIRED IN LOWER-RISK MDS WHEN COMPARED WITH REFERENCE POPULATIONS AND SIGNIFICANTLY AFFECTS OVERALL SURVIVAL
- Author
-
Stauder, R., Yu, G., Koinig, K., Fenaux, P., Argiris Symeonidis, Sanz, G., Cermak, J., Mittelman, M., Hellstrom-Lindberg, E., Langemeijer, S., Holm, M. Skov, Madry, K., Malcovati, L., Tatic, A., Germing, U., Savic, A., Efficace, F., Smith, A., Bowen, D., and Witte, T.
32. Oral Iptacopan Monotherapy in Paroxysmal Nocturnal Hemoglobinuria.
- Author
-
de Latour, R. Peffault, Roth, A., Kulasekararaj, A. G., Han, B., Scheinberg, P., Maciejewski, J. P., Ueda, Y., de Castro, C. M., Di Bona, E., Fu, R., Zhang, L., Griffin, M., Langemeijer, S. M. C., Panse, J., Schrezenmeier, H., Barcellini, W., Mauad, V. A. Q., Schafhausen, P., Tavitian, S., and Beggiato, E.
- Subjects
- *
PAROXYSMAL hemoglobinuria , *CLINICAL trials , *COMPLEMENT inhibition , *HEMOLYTIC anemia , *LACTATE dehydrogenase - Abstract
Persistent hemolytic anemia and a lack of oral treatments are challenges for patients with paroxysmal nocturnal hemoglobinuria who have received anti-C5 therapy or have not received complement inhibitors. Iptacopan, a first-in-class oral factor B inhibitor, has been shown to improve hemoglobin levels in these patients. METHODS In two phase 3 trials, we assessed iptacopan monotherapy over a 24-week period in patients with hemoglobin levels of less than 10 g per deciliter. In the first, anti-C5-treated patients were randomly assigned to switch to iptacopan or to continue anti-C5 therapy. In the second, single-group trial, patients who had not received complement inhibitors and who had lactate dehydrogenase (LDH) levels more than 1.5 times the upper limit of the normal range received iptacopan. The two primary end points in the first trial were an increase in the hemoglobin level of at least 2 g per deciliter from baseline and a hemoglobin level of at least 12 g per deciliter, each without red-cell transfusion; the primary end point for the second trial was an increase in hemoglobin level of at least 2 g per deciliter from baseline without red-cell transfusion. RESULTS In the first trial, 51 of the 60 patients who received iptacopan had an increase in the hemoglobin level of at least 2 g per deciliter from baseline, and 42 had a hemoglobin level of at least 12 g per deciliter, each without red-cell transfusions; none of the 35 anti-C5-treated patients attained the end-point levels. In the second trial, 31 of 33 patients had an increase in the hemoglobin level of at least 2 g per deciliter from baseline without red-cell transfusion. In the first trial, 59 of the 62 patients who received iptacopan and 14 of the 35 anti-C5-treated patients received no transfusion; none of the patients in the second trial received transfusions. Treatment with iptacopan increased hemoglobin levels, reduced fatigue, reduced reticulocyte and bilirubin levels, and resulted in mean LDH levels that were less than 1.5 times the upper limit of the normal range. Headache was the most frequent adverse event with iptacopan. CONCLUSIONS Iptacopan treatment improved hematologic and clinical outcomes in anti-C5-treated patients with persistent anemia -- in whom iptacopan showed superiority to anti-C5 therapy -- and in patients who had not received complement inhibitors. (Funded by Novartis; APPLY-PNH ClinicalTrials.gov number, NCT04558918; APPOINT-PNH ClinicalTrials.gov number, NCT04820530.) [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
33. Incidence and Clinical Impact of TET2Mutations in Acute Myeloid Leukemia Patients Treated within the EORTC/GIMEMA AML-12/06991 AML Trial.
- Author
-
Aslanyan, M. G, Langemeijer, S. M.C., Cilloni, D., Saglio, G., Marie, JP, Tang, R., Labar, B., Zadro, R., Batinic, D., Amadori, S., Lo Coco, F., Scheele, T., Kroeze, L., Massop, M., van Hoogen, P., Stevens, E., Muus, P., Suciu, S., Baila, L., Marijt, E. W.A., Willemze, R., de Witte, T., van der Reijden, B., and Jansen, J.H.
- Abstract
Abstract 2609
- Published
- 2009
- Full Text
- View/download PDF
34. Survival and quality of life in patients with lower risk myelodysplastic syndromes exposed to erythropoiesis-stimulating agents: an observational cohort study.
- Author
-
Garelius HKG, Bagguley T, Taylor A, Fenaux P, Bowen D, Symeonidis A, Mittelmann M, Stauder R, Čermák J, Sanz G, Langemeijer S, Malcovati L, Germing U, Sanhes L, d'Aveni M, Culligan D, Kotsianidis I, Koinig KA, van Marrewijk C, Crouch S, deWitte T, Smith A, and Hellström-Lindberg E
- Subjects
- Humans, Female, Male, Aged, Middle Aged, Erythrocyte Transfusion adverse effects, Registries, Aged, 80 and over, Cohort Studies, Longitudinal Studies, Quality of Life, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes mortality, Myelodysplastic Syndromes therapy, Myelodysplastic Syndromes complications, Hematinics therapeutic use
- Abstract
Background: In our previous study on erythropoiesis-stimulating agent (ESA) treatment in lower risk myelodysplastic syndromes from the European MDS (EUMDS) Registry, we showed that patients treated with ESAs had longer survival compared with patients who receive red blood cell transfusion (RBCT). In this study, with a longer follow up time and more patients included, we aimed to assess long-term effects on survival and health-related quality of life (HRQoL) of exposure to ESAs with or without RBCT in patients with lower risk myelodysplastic syndromes., Methods: The EUMDS Registry is a non-interventional, longitudinal, real-world registry prospectively enrolling newly diagnosed patients older than 18 years with lower risk (International Prognostic Scoring System low or intermediate-1) myelodysplastic syndromes from 16 European countries and Israel. The analysis was restricted to patients with haemoglobin concentrations less than 100 g/L enrolled between Jan 1, 2008, and July 1, 2019, with last censoring of data on Dec 31, 2021. Patient management was recorded every 6 months, including treatment, transfusions, and HRQoL. ESA treatment followed local guidelines. The patients were separated into four groups at each study visit: no ESA or RBCT, ESA only, ESA plus RBCT, and RBCT only. The data were analysed longitudinally over time according to ESA and RBCT status during each 6-month interval, using propensity score matching. The main outcomes were median overall survival and leukaemia-free survival, and HRQoL. This study is registered with ClinicalTrials.gov, NCT00600860, as is ongoing., Findings: 2448 patients (the ESA-unexposed group [n=1265] and ESA-exposed group [n=1183]) were diagnosed before July 1, 2019; 1520 (62·1%) were male and 928 (37·9%) were female. Median follow-up time was 3·9 years (IQR 1·6-6·5). After applying eligibility criteria and propensity matching, there were 426 patients in the ESA-unexposed group and 744 patients in the ESA-exposed group. Median overall survival in the ESA exposed group was 44·9 months (95% CI 40·2-50·5) compared with 34·8 months (28·6-39·2) in the ESA unexposed group; the absolute difference was 10·1 months (95% CI 2·2-18·0; hazard ratio [HR] 0·70 [95% CI 0·59-0·83]; p<0·0001). Patients without RBCT in the presence or absence of ESA exposure maintained significantly better HRQoL than those with RBCT, irrespective of ESA exposure (linear mixed effect model of EQ-5d-3L index score, RBCT coefficient -0·04 [95% CI -0·06 to 0·03], p<0·0001; linear mixed effect model of VAS, -4·57 [-6·02 to -3·13], p<0·0001)., Interpretation: ESA treatment in patients with lower risk myelodysplastic syndromes significantly improves overall survival when started before or early after the onset of regular transfusion therapy. Avoiding RBCT is associated with significantly better HRQoL., Funding: H2020 European Research Council, Novartis Pharmacy B V Oncology Europe, Amgen, BMS/Celgene International, Janssen Pharmaceutica, Takeda Pharmaceuticals International, and Gilead Sciences., Competing Interests: Declaration of interests RS reports honoraria from BMS for the MDS Right Horizon 2020 Project; financial support for meetings from Celgene/BMS, Lilly, Teva, AbbVie, and AHOP; advisory board participation for BMS and Otsuka; and is Servier President of Verein Senioren Krebshilfe. CvM and TdW work for the EUMDS Registry, which is supported by an educational grant from Novartis Pharmacy BV Oncology Europe, Amgen, BMS, Janssen Pharmaceutical, Takeda Pharmaceutical, Gilead Sciences, and a European commission H2020 Grant. AS is supported in part by the National Institute for Health and Care Research (NIHR) Leeds Biomedical Research Centre (NIHR203331). HKGG, TB, AT, PF, DB, ArS, JC, SL, LM, UG, LS, MdA, DC, KAK, SCr, and EHL declare no competing interests., (Copyright © 2025 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
35. Somatic mutations and DNA methylation identify a subgroup of poor prognosis within lower-risk myelodysplastic syndromes.
- Author
-
Rombaut D, Sandmann S, Tekath T, Crouch S, de Graaf AO, Smith A, Painter D, Kosmider O, Tobiasson M, Lennartsson A, van der Reijden BA, Park S, D'Aveni M, Slama B, Clappier E, Fenaux P, Adès L, van de Loosdrecht A, Langemeijer S, Symeonidis A, Čermák J, Preudhomme C, Savic A, Germing U, Stauder R, Bowen D, van Marrewijk C, Bernard E, de Witte T, Varghese J, Hellström-Lindberg E, Dugas M, Martens J, Malcovati L, Jansen JH, and Fontenay M
- Abstract
Lower risk (LR) myelodysplastic syndromes (MDS) are heterogeneous hematopoietic stem and progenitor disorders caused by the accumulation of somatic mutations in various genes including epigenetic regulators that may produce convergent DNA methylation patterns driving specific gene expression profiles. The integration of genomic, epigenomic, and transcriptomic profiling has the potential to spotlight distinct LR-MDS categories on the basis of pathophysiological mechanisms. We performed a comprehensive study of somatic mutations and DNA methylation in a large and clinically well-annotated cohort of treatment-naive patients with LR-MDS at diagnosis from the EUMDS registry (ClinicalTrials.gov.NCT00600860). Unsupervised clustering analyses identified six clusters based on genetic profiling that concentrate into four clusters on the basis of genome-wide methylation profiling with significant overlap between the two clustering modes. The four methylation clusters showed distinct clinical and genetic features and distinct methylation landscape. All clusters shared hypermethylated enhancers enriched in binding motifs for ETS and bZIP (C/EBP) transcription factor families, involved in the regulation of myeloid cell differentiation. By contrast, one cluster gathering patients with early leukemic evolution exhibited a specific pattern of hypermethylated promoters and, distinctly from other clusters, the upregulation of AP-1 complex members FOS/FOSL2 together with the absence of hypermethylation of their binding motif at target gene enhancers, which is of relevance for leukemic initiation. Among MDS patients with lower-risk IPSS-M, this cluster displayed a significantly inferior overall survival ( p < 0.0001). Our study showed that genetic and DNA methylation features of LR-MDS at early stages may refine risk stratification, therefore offering the frame for a precocious therapeutic intervention., Competing Interests: The authors declare no conflict of interest., (© 2025 The Author(s). HemaSphere published by John Wiley & Sons Ltd on behalf of European Hematology Association.)
- Published
- 2025
- Full Text
- View/download PDF
36. Comparative clinical efficacy and safety of biosimilar ABP 959 and eculizumab reference product in patients with paroxysmal nocturnal hemoglobinuria.
- Author
-
Kulasekararaj A, Lanza F, Arvanitakis A, Langemeijer S, Chonat S, Tombak A, Hanes V, Cao J, Miller MJ, Colbert A, Shander B, Mytych DT, Chow V, and Henary H
- Subjects
- Humans, Male, Female, Middle Aged, Adult, Double-Blind Method, L-Lactate Dehydrogenase blood, Aged, Treatment Outcome, Hemolysis drug effects, Hemoglobinuria, Paroxysmal drug therapy, Antibodies, Monoclonal, Humanized therapeutic use, Antibodies, Monoclonal, Humanized pharmacokinetics, Antibodies, Monoclonal, Humanized adverse effects, Antibodies, Monoclonal, Humanized administration & dosage, Biosimilar Pharmaceuticals therapeutic use, Biosimilar Pharmaceuticals pharmacokinetics, Biosimilar Pharmaceuticals adverse effects, Cross-Over Studies
- Abstract
ABP 959 is a biosimilar to the eculizumab reference product (RP), which is approved for the treatment of patients with paroxysmal nocturnal hemoglobinuria (PNH). This multicenter, randomized, double-blind, active-controlled, two-period crossover study randomized eculizumab RP-treated patients with PNH to one of two treatment sequences (ABP 959/eculizumab RP or eculizumab RP/ABP 959) to evaluate the clinical similarity of ABP 959 when compared with eculizumab RP. This study evaluated the efficacy of ABP 959 when compared with eculizumab RP based on control of intravascular hemolysis as measured by lactate dehydrogenase (LDH) and by the time-adjusted area under the effect curve of LDH. Secondary outcomes included safety, pharmacokinetics, and immunogenicity. Forty-two patients were randomized (20 in the ABP 959/eculizumab RP group and 22 in the eculizumab RP/ABP 959 group) across 25 centers. Similarity of efficacy was established by a ratio of geometric least squares means of LDH (ABP 959/eculizumab RP) of 1.0628, with a one-sided 97.5% upper CI of 1.1576 at week 27, and a geometric means ratio of time-adjusted area under the effect curve (ABP 959 vs. eculizumab RP) of LDH of 0.981, with a 90% CI of 0.9403-1.0239 from week 13 to 27, week 39 to 53, and week 65 to 79. All secondary efficacy endpoints were comparable between treatment groups. No new safety concerns were identified. The results of this study in patients with PNH, along with previously demonstrated similarity of analytical, nonclinical, and clinical pharmacokinetics and pharmacodynamics in healthy volunteers support a demonstration of no clinically meaningful differences between ABP 959 and eculizumab RP. Clinical Trial Registration: NCT03818607., (© 2024 The Author(s). American Journal of Hematology published by Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
37. Inflammatory profile of lower risk myelodysplastic syndromes.
- Author
-
Topping J, Taylor A, Nadat F, Crouch S, Ibbotson A, Čermák J, Symeonidis A, Tatic A, Langemeijer S, Hellström-Lindberg E, Culligan D, Garelius HG, Ashcroft J, Nga E, Parker J, Kolade S, McDermott MF, De Witte T, Bowen D, Smith A, Cargo C, and Savic S
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Biomarkers blood, Inflammation blood, Inflammasomes, NLR Family, Pyrin Domain-Containing 3 Protein blood, Adult, Case-Control Studies, Aged, 80 and over, Myelodysplastic Syndromes blood, Cytokines blood
- Abstract
The precise link between inflammation and pathogenesis of myelodysplastic syndrome (MDS) is yet to be fully established. We developed a novel method to measure ASC/NLRP3 protein specks which are specific for the NLRP3 inflammasome only. We combined this with cytokine profiling to characterise various inflammatory markers in a large cohort of patients with lower risk MDS in comparison to healthy controls and patients with defined autoinflammatory disorders (AIDs). The ASC/NLRP3 specks were significantly elevated in MDS patients compared to healthy controls (p < 0.001) and these levels were comparable to those found in patients with AIDs. The distribution of protein specks positive only for ASC was different to ASC/NLRP3 ones suggesting that other ASC-containing inflammasome complexes might be important in the pathogenesis of MDS. Patients with MDS-SLD had the lowest levels of interleukin (IL)-1β, tumour necrosis factor (TNF), IL-23, IL-33, interferon (IFN) γ and IFN-α2, compared to other diagnostic categories. We also found that inflammatory cytokine TNF was positively associated with MDS progression to a more aggressive form of disease and IL-6 and IL-1β with time to first red blood cell transfusion. Our study shows that there is value in analysing inflammatory biomarkers in MDS, but their diagnostic and prognostic utility is yet to be fully validated., (© 2024 The Authors. British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
38. Identification and surveillance of rare relapse-initiating stem cells during complete remission after transplantation.
- Author
-
Dimitriou M, Mortera-Blanco T, Tobiasson M, Mazzi S, Lehander M, Högstrand K, Karimi M, Walldin G, Jansson M, Vonlanthen S, Ljungman P, Langemeijer S, Yoshizato T, Hellström-Lindberg E, Woll PS, and Jacobsen SEW
- Subjects
- Humans, Transplantation, Homologous, Retrospective Studies, Neoplasm Recurrence, Local, Pathologic Complete Response, Chronic Disease, Neoplastic Stem Cells pathology, Recurrence, Neoplasm, Residual diagnosis, Neoplasm, Residual pathology, Hematopoietic Stem Cell Transplantation, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute therapy
- Abstract
Abstract: Relapse after complete remission (CR) remains the main cause of mortality after allogeneic stem cell transplantation for hematological malignancies and, therefore, improved biomarkers for early prediction of relapse remains a critical goal toward development and assessment of preemptive relapse treatment. Because the significance of cancer stem cells as a source of relapses remains unclear, we investigated whether mutational screening for persistence of rare cancer stem cells would enhance measurable residual disease (MRD) and early relapse prediction after transplantation. In a retrospective study of patients who relapsed and patients who achieved continuous-CR with myelodysplastic syndromes and related myeloid malignancies, combined flow cytometric cell sorting and mutational screening for persistence of rare relapse-initiating stem cells was performed in the bone marrow at multiple CR time points after transplantation. In 25 CR samples from 15 patients that later relapsed, only 9 samples were MRD-positive in mononuclear cells (MNCs) whereas flowcytometric-sorted hematopoietic stem and progenitor cells (HSPCs) were MRD-positive in all samples, and always with a higher variant allele frequency than in MNCs (mean, 97-fold). MRD-positivity in HSPCs preceded MNCs in multiple sequential samples, in some cases preceding relapse by >2 years. In contrast, in 13 patients in long-term continuous-CR, HSPCs remained MRD-negative. Enhanced MRD sensitivity was also observed in total CD34+ cells, but HSPCs were always more clonally involved (mean, 8-fold). In conclusion, identification of relapse-initiating cancer stem cells and mutational MRD screening for their persistence consistently enhances MRD sensitivity and earlier prediction of relapse after allogeneic stem cell transplantation., (© 2024 American Society of Hematology. Published by Elsevier Inc. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
39. Determinants of low health-related quality of life in patients with myelodysplastic syndromes: EUMDS Registry study.
- Author
-
Stojkov I, Conrads-Frank A, Rochau U, Arvandi M, Koinig KA, Schomaker M, Mittelman M, Fenaux P, Bowen D, Sanz GF, Malcovati L, Langemeijer S, Germing U, Madry K, Guerci-Bresler A, Culligan DJ, Kotsianidis I, Sanhes L, Mills J, Puntscher S, Schmid D, van Marrewijk C, Smith A, Efficace F, de Witte T, Stauder R, and Siebert U
- Subjects
- Aged, Female, Humans, Middle Aged, Comorbidity, Cross-Sectional Studies, Myelodysplastic Syndromes diagnosis, Myelodysplastic Syndromes epidemiology, Myelodysplastic Syndromes therapy, Quality of Life
- Abstract
Patients with myelodysplastic syndromes (MDS) frequently experience a significant symptom burden, which reduces health-related quality of life (HRQoL). We aimed to identify determinants of low HRQoL in patients recently diagnosed with MDS, for guiding early intervention strategies. We evaluated longitudinal data in 2205 patients with MDS during their first year after diagnosis. Median values of EQ-5D 3-level (EQ-5D-3L) index (0.78) and visual analog scale (VAS) score (0.70) were used as thresholds for low HRQoL. In addition, the 5 dimensions of EQ-5D-3L were analyzed for impairments (any level vs "no problem" category). After multiple imputation of missing values, we used generalized estimating equations (GEE) to estimate odds ratios (OR) for univariable determinant screening (P < .15), and to subsequently derive multivariable models for low HRQoL with 95% confidence intervals (CI). Multivariable GEE analysis showed the following independent determinants (OR, 95% CI) for low EQ-5D index: increased age (60-75 years: 1.33, 1.01-1.75; >75: 1.84, 1.39-2.45), female sex (1.70, 1.43-2.03), high serum ferritin level (≥1000 vs ≤300 μg/L: 1.41, 1.06-1.87), comorbidity burden (per unit: 1.11, 1.02-1.20), and reduced Karnofsky performance status (KPS, per 10 units: 0.62, 0.58-0.67). For low VAS score, additional determinants were transfusion dependence (1.53, 1.03-2.29), low hemoglobin <10 g/dL (1.34, 1.12-1.61), and high body mass index (≥30 vs 23-29.9 kg/m2: 1.26, 1.02-1.57). Sex, KPS, comorbidity burden, hemoglobin count, and transfusion burden were determinants for all EQ-5D dimensions. Low HRQoL is determined by multiple factors, which should be considered in the management and shared decision making of patients with MDS. This trial was registered at www.clinicaltrials.gov as #NCT00600860., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
40. Raising the standards of patient-centered outcomes research in myelodysplastic syndromes: Clinical utility and validation of the subscales of the QUALMS from the MDS-RIGHT project.
- Author
-
Efficace F, Koinig K, Cottone F, Bowen D, Mittelman M, Sommer K, Langemeijer S, Culligan D, Filanovsky K, Storck M, Smith A, van Marrewijk C, Dugas M, Stojkov I, Siebert U, de Witte T, and Stauder R
- Subjects
- Humans, Aged, Quality of Life, Prospective Studies, Patient Outcome Assessment, Myelodysplastic Syndromes therapy, Myelodysplastic Syndromes drug therapy, Anemia
- Abstract
Background: Clinical decision-making for patients with myelodysplastic syndromes (MDS) is challenging, and both disease and treatment effects heavily impact health-related quality of life (HRQoL) of these patients. Therefore, disease-specific HRQoL measures can be critical to harness the patient voice in MDS research., Methods: We report a prospective international validation study of the Quality of Life in Myelodysplasia Scale (QUALMS) with a main focus on providing information on the psychometric characteristics of its three subscales: physical burden (QUALMS-P), emotional burden (QUALMS-E), and benefit finding (QUALMS-BF). The analysis is based on patients enrolled from three European countries and Israel, participating to the MDS-RIGHT Project. The scale structure and psychometric properties of the QUALMS were assessed., Results: Overall, 270 patients with a median age of 74 years were analyzed and the majority of them (60.3%) had a low MDS-Comorbidity Index score. Results of the confirmatory factor analysis supported the underlying scale structure of the QUALMS, which, in addition to a total score, includes three subscales: QUALMS-P, QUALMS-E, and the QUALMS-BF. The QUALMS-P exhibited the highest Cronbach's alpha coefficients. Discriminant validity analysis indicated good results with the QUALMS-P and QUALMS-E distinguishing between patients with different performance status, comorbidity, anemia, and transfusion dependency status. No floor and ceiling effects were observed. Responsiveness to change analysis supported the validity of the measure. Patients with a hemoglobin (Hb) level of <11 g/dL at study entry, who subsequently showed an improvement in their Hb levels, also reported a mean score change of 9 and 8 points (scales ranging between 0 and 100) in the expected direction of the QUALMS-E and QUALMS-P, respectively., Conclusions: Our study provides additional validation data on the QUALMS from the international MDS-RIGHT Project. The use of this disease-specific HRQoL measure may contribute to raise quality standards of patient-centered outcomes research in MDS., (© 2022 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
41. Cause of death and excess mortality in patients with lower-risk myelodysplastic syndromes (MDS): A report from the European MDS registry.
- Author
-
Mądry K, Lis K, Fenaux P, Bowen D, Symeonidis A, Mittelman M, Stauder R, Čermák J, Sanz G, Hellström-Lindberg E, Langemeijer S, Malcovati L, Germing U, Holm MS, Guerci-Bresler A, Culligan D, Sanhes L, Kotsianidis I, van Marrewijk C, Crouch S, de Witte T, and Smith A
- Subjects
- Humans, Cause of Death, Disease Progression, Registries, Myelodysplastic Syndromes, Cardiovascular Diseases, Leukemia, Myeloid, Acute
- Abstract
Information on causes of death (CoDs) and the impact of myelodysplastic syndromes (MDS) on survival in patients with lower-risk MDS (LR-MDS) is limited. A better understanding of the relationship between disease characteristics, clinical interventions and CoDs may improve outcomes of patients with LR-MDS. We prospectively collected data on patients with LR-MDS in the European MDS registry from 2008 to 2019. Clinical, laboratory and CoDs data were obtained. To examine MDS-specific survival, relative survival (RS) was estimated using national life tables. Of 2396 evaluated subjects, 900 died (median overall survival [OS]: 4.7 years; median follow-up: 3.5 years). The most common CoDs were acute myeloid leukaemia/MDS (20.1%), infection (17.8%) and cardiovascular disease (CVD; 9.8%). Patients with isolated del(5q) and with red cell transfusion needed during the disease course, had a higher risk of fatal CVD. The 5-year OS was 47.3% and the 5-year RS was 59.6%, indicating that most patients died due to their underlying MDS. Older patients (aged >80 years) and the lowest-risk patients were more likely to die from competing causes. This study shows that MDS and its related complications play crucial role in the outcome of patients with LR-MDS., (© 2022 British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
42. Long-term safety and efficacy of ravulizumab in patients with paroxysmal nocturnal hemoglobinuria: 2-year results from two pivotal phase 3 studies.
- Author
-
Kulasekararaj AG, Griffin M, Langemeijer S, Usuki K, Kulagin A, Ogawa M, Yu J, Mujeebuddin A, Nishimura JI, Lee JW, and Peffault de Latour R
- Subjects
- Antibodies, Monoclonal, Humanized, Complement C5, Complement Inactivating Agents adverse effects, Hemolysis, Humans, Hemoglobinuria, Paroxysmal diagnosis, Hemoglobinuria, Paroxysmal drug therapy
- Abstract
Objectives: The complement component 5 (C5) inhibitor ravulizumab demonstrated non-inferiority to eculizumab following 26 weeks of treatment in complement inhibitor-naïve and complement inhibitor-experienced patients with paroxysmal nocturnal hemoglobinuria (PNH; studies 301 and 302, respectively). This study aims to describe the results of both studies from 27 weeks to 2 years., Methods: Patients (N = 441) continued to receive ravulizumab throughout the extension period. Efficacy endpoints included lactate dehydrogenase (LDH) normalization, transfusion avoidance and fatigue score (FACIT-F). Safety analyses were also performed., Results: From 27 weeks to 2 years, improvements in LDH levels were maintained in both study populations. Transfusion avoidance was maintained in 81.9% (study 301) and 85.6% (study 302) of patients, and FACIT-F scores remained stable. Ravulizumab was well tolerated, and the incidence of adverse events (AEs) were similar between patients of both studies. Incidence of serious AEs deemed related to ravulizumab treatment was low (<3%)., Conclusions: This study reports, to date, the longest period of follow-up in over 400 patients with PNH treated with ravulizumab (662 patient-years). Long-term, ravulizumab demonstrated durable efficacy and was well tolerated, highlighting the importance of C5 inhibitors as the mainstay of PNH treatment., (© 2022 Alexion, AstraZeneca Rare Disease. European Journal of Haematology published by John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
43. A predictive algorithm using clinical and laboratory parameters may assist in ruling out and in diagnosing MDS.
- Author
-
Oster HS, Crouch S, Smith A, Yu G, Abu Shrkihe B, Baruch S, Kolomansky A, Ben-Ezra J, Naor S, Fenaux P, Symeonidis A, Stauder R, Cermak J, Sanz G, Hellström-Lindberg E, Malcovati L, Langemeijer S, Germing U, Holm MS, Madry K, Guerci-Bresler A, Culligan D, Sanhes L, Mills J, Kotsianidis I, van Marrewijk C, Bowen D, de Witte T, and Mittelman M
- Subjects
- Algorithms, Bone Marrow Examination, Humans, Laboratories, Bone Marrow Diseases, Myelodysplastic Syndromes diagnosis
- Abstract
We present a noninvasive Web-based app to help exclude or diagnose myelodysplastic syndrome (MDS), a bone marrow (BM) disorder with cytopenias and leukemic risk, diagnosed by BM examination. A sample of 502 MDS patients from the European MDS (EUMDS) registry (n > 2600) was combined with 502 controls (all BM proven). Gradient-boosted models (GBMs) were used to predict/exclude MDS using demographic, clinical, and laboratory variables. Area under the receiver operating characteristic curve (AUC), sensitivity, and specificity were used to evaluate the models, and performance was validated using 100 times fivefold cross-validation. Model stability was assessed by repeating its fit using different randomly chosen groups of 502 EUMDS cases. AUC was 0.96 (95% confidence interval, 0.95-0.97). MDS is predicted/excluded accurately in 86% of patients with unexplained anemia. A GBM score (range, 0-1) of less than 0.68 (GBM < 0.68) resulted in a negative predictive value of 0.94, that is, MDS was excluded. GBM ≥ 0.82 provided a positive predictive value of 0.88, that is, MDS. The diagnosis of the remaining patients (0.68 ≤ GBM < 0.82) is indeterminate. The discriminating variables: age, sex, hemoglobin, white blood cells, platelets, mean corpuscular volume, neutrophils, monocytes, glucose, and creatinine. A Web-based app was developed; physicians could use it to exclude or predict MDS noninvasively in most patients without a BM examination. Future work will add peripheral blood cytogenetics/genetics, EUMDS-based prospective validation, and prognostication., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
44. A phase I clinical trial to study the safety of treatment with tipifarnib combined with bortezomib in patients with advanced stages of myelodysplastic syndrome and oligoblastic acute myeloid leukemia.
- Author
-
Muus P, Langemeijer S, van Bijnen S, Blijlevens N, and de Witte T
- Subjects
- Aged, Bortezomib administration & dosage, Dose-Response Relationship, Drug, Female, Humans, Male, Maximum Tolerated Dose, Middle Aged, Quinolones administration & dosage, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols adverse effects, Bortezomib adverse effects, Leukemia, Myeloid, Acute drug therapy, Myelodysplastic Syndromes drug therapy, Quinolones adverse effects
- Abstract
Purpose: To determine the safety of tipifarnib in combination with escalating doses of bortezomib and to determine the maximum tolerated dose in patients with untreated high-risk MDS and oligoblastic acute myeloid leukemia, who were not eligible for intensive therapy., Experimental Design: In a "3 + 3″ design, patients received fixed doses of tipifarnib 200 mg bid (days 1-21) and escalating doses of bortezomib (days 8, 15, 22) every 4 weeks in 4-6 cycles., Results: The combination was tolerated well by the 11 patients in this study without reaching the maximum tolerated dose. Myelosuppression was the most frequent side effect, but usually of short duration. Interestingly a complete response with or without complete count recovery was observed in three patients and three additional patients had stable disease. The median duration of overall survival was 449 days. Two patients were still alive at 4.0 and 4.3 years, including one patient in continuing CR., Conclusions: The combination of tipifarnib and bortezomib was tolerated well and appeared to have clinical activity in patients with high-risk MDS and AML with low counts of marrow blasts. Our results warrant further evaluation in a phase II study., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
45. Placental disposition of eculizumab, C5 and C5-eculizumab in two pregnancies of a woman with paroxysmal nocturnal haemoglobinuria.
- Author
-
Eliesen GAM, van Drongelen J, van den Broek PHH, Sarlea A, van der Heijden OWH, Langemeijer S, Greupink R, Volokhina EB, and Russel FGM
- Subjects
- Antibodies, Monoclonal, Humanized, Female, Humans, Infant, Newborn, Placenta, Pregnancy, Hemoglobinuria, Paroxysmal drug therapy
- Abstract
Eculizumab is known to cross the placenta to a limited degree, but recently therapeutic drug levels in cord blood were found in a single case. We report maternal, cord and placental levels of unbound eculizumab, C5 and C5-eculizumab in two pregnancies of a paroxysmal nocturnal haemoglobinuria patient who received 900 mg eculizumab every 2 weeks. In both pregnancies, cord blood concentrations of unbound eculizumab were below 4 μg/mL, while C5-eculizumab levels were 22 and 26 μg/mL, suggesting that a considerable fraction of C5 was blocked in the newborn. Concentrations in each placenta of unbound eculizumab were 41 ± 3 and 45 ± 4 μg/g tissue, of C5-eculizumab 19 ± 2 and 32 ± 3 μg/g, and of C5 20 ± 3 and 30 ± 2 μg/g (mean ± SD, in three tissue samples per placenta). Placental levels of unbound eculizumab were higher than those of C5-eculizumab complexes, while maternal concentrations were approximately equal, suggesting selective transport of unbound eculizumab across the placenta., (© 2020 The Authors. British Journal of Clinical Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.)
- Published
- 2021
- Full Text
- View/download PDF
46. One-year outcomes from a phase 3 randomized trial of ravulizumab in adults with paroxysmal nocturnal hemoglobinuria who received prior eculizumab.
- Author
-
Kulasekararaj AG, Hill A, Langemeijer S, Wells R, González Fernández FA, Gaya A, Ojeda Gutierrez E, Piatek CI, Mitchell L, Usuki K, Bosi A, Brodsky RA, Ogawa M, Yu J, Ortiz S, Röth A, Lee JW, and Peffault de Latour R
- Subjects
- Adult, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Blood Transfusion, Combined Modality Therapy, Complement C5 immunology, Complement C5 metabolism, Complement Inactivating Agents administration & dosage, Complement Inactivating Agents adverse effects, Female, Hemoglobinuria, Paroxysmal blood, Hemoglobinuria, Paroxysmal diagnosis, Hemolysis, Humans, Male, Molecular Targeted Therapy, Quality of Life, Retreatment, Treatment Outcome, Antibodies, Monoclonal, Humanized therapeutic use, Complement Inactivating Agents therapeutic use, Hemoglobinuria, Paroxysmal drug therapy
- Abstract
Ravulizumab every 8 weeks showed non-inferiority to eculizumab every 2 weeks in a 26-week, phase 3, randomized controlled trial in adults with paroxysmal nocturnal hemoglobinuria (PNH) who were clinically stable on eculizumab (NCT03056040). We report results from the first 26 weeks of the extension period in which patients continued ravulizumab (n = 96) or switched from eculizumab to ravulizumab (n = 95). At week 52, mean (SD) lactate dehydrogenase levels increased 8.8% (29%) with ravulizumab-ravulizumab and 5.8% (27%) with eculizumab-ravulizumab from primary evaluation period baseline. During the extension period, four patients (ravulizumab-ravulizumab, n = 3; eculizumab-ravulizumab, n = 1) experienced breakthrough hemolysis, but none associated with serum free C5 ≥ 0.5 μg/mL. Mean Functional Assessment of Chronic Illness Therapy (FACIT)-Fatigue scores remained stable through week 52. During the extension period, proportions of patients avoiding transfusion remained stable (ravulizumab-ravulizumab, 86.5%; eculizumab-ravulizumab, 83.2%); 81.2% and 81.1%, respectively, had stabilized hemoglobin. All patients maintained serum free C5 levels < 0.5 μg/mL. Adverse events were generally similar between groups, and rates were lower in the extension period. Adults with PNH on stable eculizumab therapy who received ravulizumab over 52 weeks experienced durable efficacy, with consistent efficacy in patients who received eculizumab during the primary evaluation period and then switched to ravulizumab. Ravulizumab was well tolerated., (© 2020 The Authors. European Journal of Haematology published by John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
47. Paroxysmal nocturnal hemoglobinuria caused by CN-LOH of constitutional PIGB mutation and 70-kbp microdeletion on 15q.
- Author
-
Langemeijer S, Schaap C, Preijers F, Jansen JH, Blijlevens N, Inoue N, Muus P, Kinoshita T, and Murakami Y
- Subjects
- Animals, CHO Cells, Cricetinae, Cricetulus, Female, Humans, Loss of Heterozygosity, Mannosyltransferases, Membrane Proteins genetics, Mutation, Hemoglobinuria, Paroxysmal genetics
- Abstract
Paroxysmal nocturnal hemoglobinuria (PNH) is a rare hematopoietic stem cell (HSC) disorder characterized by defective synthesis of the glycosylphosphatidylinositol (GPI) anchors as a result of somatic mutations in the X-linked PIGA gene. The disease is acquired. No constitutional PNH has been described. Here, we report familial PNH associated with unusual inflammatory symptoms. Genetic analysis revealed a germline heterozygous PIGB mutation on chromosome 15 without mutations in PIGA or any of the other genes involved in GPI biosynthesis. In vitro data confirmed that transfection of the mutant PIGB could not restore the surface expression of GPI-anchored proteins (APs) in PIGB-deficient Chinese hamster ovary cells. Homozygosity was caused by copy number-neutral loss of heterozygosity (CN-LOH) of the germline PIGB mutation, leading to deficient expression of GPI-APs in the affected blood cells of the index patient and her mother. The somatic event leading to homozygosity of the germline mutant PIGB gene involved a 70-kbp microdeletion of chromosome 15q containing the TM2D3 and TARSL2 genes, which was implicated in chromosome 15q mosaicism. Interestingly, we detected the deletion in both the patient and her mother. A sister of the mother, who carried the same germline PIGB mutation but without this microdeletion involving TM2D3 and TARSL2, did not have a PNH clone or CN-LOH. In conclusion, we describe PNH caused by CN-LOH of a germline heterozygous PIGB mutation in a patient and her mother and hypothesize that the 70-kbp microdeletion may have contributed to the PNH clone in both., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
48. Novel dynamic outcome indicators and clinical endpoints in myelodysplastic syndrome; the European LeukemiaNet MDS Registry and MDS-RIGHT project perspective.
- Author
-
De Witte T, Malcovati L, Fenaux P, Bowen D, Symeonidis A, Mittelman M, Stauder R, Sanz G, Čermák J, Langemeijer S, Hellström-Lindberg E, Germing U, Skov Holm M, Mądry K, Tatic A, Medina Almeida A, Savic A, Mandac Rogulj I, Itzykson R, Hoeks M, Gravdahl Garelius H, Culligan D, Kotsianidis I, Ades L, Van de Loosdrecht AA, Van Marrewijk C, Yu G, Crouch S, and Smith A
- Subjects
- Humans, Registries, Myelodysplastic Syndromes diagnosis, Myelodysplastic Syndromes epidemiology, Myelodysplastic Syndromes therapy
- Published
- 2020
- Full Text
- View/download PDF
49. Eculizumab impairs Neisseria meningitidis serogroup B killing in whole blood despite 4CMenB vaccination of PNH patients.
- Author
-
Langereis JD, van den Broek B, Franssen S, Joosten I, Blijlevens NMA, de Jonge MI, and Langemeijer S
- Subjects
- Antibodies, Monoclonal, Humanized, Humans, Serogroup, Vaccination, Hemoglobinuria, Paroxysmal drug therapy, Meningococcal Infections drug therapy, Meningococcal Infections prevention & control, Meningococcal Vaccines, Neisseria meningitidis
- Abstract
Complement C5 inhibitor eculizumab has a great impact on the treatment of patients with paroxysmal nocturnal hemoglobinuria (PNH). However, this treatment success has a major drawback: a substantially increased susceptibility for life-threatening Neisseria meningitidis infections. Therefore, N meningitidis vaccination is strongly advised before initiating complement C5-blocking therapy. In this study, we show that the multicomponent N meningitidis serogroup B (4CMenB) vaccination of PNH patients treated with eculizumab results in a significant increase in anti-N meningitidis serogroup B (MenB) plasma immunoglobulin G (IgG) levels. Anti-MenB IgG was able to bind to the bacterial surface and initiate complement activation; however, inhibition of the membrane attack complex formation completely blocked whole blood-mediated killing of MenB. This would suggest that, despite 4CMenB vaccination, PNH patients taking C5 inhibitors are not sufficiently protected against MenB infection, which is in line with the fact that vaccinated PNH patients still experience meningococcal infections., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
50. Impact of treatment with iron chelation therapy in patients with lower-risk myelodysplastic syndromes participating in the European MDS registry.
- Author
-
Hoeks M, Yu G, Langemeijer S, Crouch S, de Swart L, Fenaux P, Symeonidis A, Čermák J, Hellström-Lindberg E, Sanz G, Stauder R, Holm MS, Mittelman M, Mądry K, Malcovati L, Tatic A, Almeida AM, Germing U, Savic A, Šimec NG, Culligan D, Itzykson R, Guerci-Bresler A, Slama B, Droste J, van Marrewijk C, van de Loosdrecht A, Blijlevens N, van Kraaij M, Bowen D, de Witte T, and Smith A
- Subjects
- Chelation Therapy, Humans, Iron therapeutic use, Iron Chelating Agents therapeutic use, Registries, Retrospective Studies, Iron Overload drug therapy, Iron Overload etiology, Myelodysplastic Syndromes drug therapy
- Abstract
Iron overload due to red blood cell (RBC) transfusions is associated with morbidity and mortality in lower-risk myelodysplastic syndrome (MDS) patients. Many studies have suggested improved survival after iron chelation therapy (ICT), but valid data are limited. The aim of this study was to assess the effect of ICT on overall survival and hematologic improvement in lower-risk MDS patients in the European MDS registry. We compared chelated patients with a contemporary, non-chelated control group within the European MDS registry, that met the eligibility criteria for starting iron chelation. A Cox proportional hazards model was used to assess overall survival (OS), treating receipt of chelation as a time-varying variable. Additionally, chelated and non-chelated patients were compared using a propensity-score matched model. Of 2,200 patients, 224 received iron chelation. The hazard ratio and 95% confidence interval for OS for chelated patients, adjusted for age, sex, comorbidity, performance status, cumulative RBC transfusions, Revised-International Prognostic Scoring System (IPSS-R), and presence of ringed sideroblasts was 0.50 (0.34-0.74). The propensity-score analysis, matched for age, sex, country, RBC transfusion intensity, ferritin level, comorbidity, performance status, and IPSS-R, and, in addition, corrected for cumulative RBC transfusions and presence of ringed sideroblasts, demonstrated a significantly improved OS for chelated patients with a hazard ratio of 0.42 (0.27-0.63) compared to non-chelated patients. Up to 39% of chelated patients reached an erythroid response. In conclusion, our results suggest that iron chelation may improve OS and hematopoiesis in transfused lower-risk MDS patients. This trial was registered at clinicaltrials.gov identifier: 00600860., (Copyright© 2020 Ferrata Storti Foundation.)
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.