1
|
Urrechaga E. Screening of Myelodysplastic Syndromes Using Research Parameters of Complete Blood Count: Automated Detection of Dysplasia. Clin Lab Med 2024; 44:387-396. [PMID: 39089745 DOI: 10.1016/j.cll.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Myelodysplastic syndromes (MDS) present with polymorphic and non-specific diagnostic features Research parametersfrom hematology analyzers may be useful to discriminate MDS-related cytopenia.Parameters such as Neu X (related to the cytoplasmic complexity) and Neu Y (related to nucleic acid content) show promise to detect dysplasia of MDS and aid to recognize MDS from cytopenias of other etiologies.
Collapse
Affiliation(s)
- Eloísa Urrechaga
- Hematology Laboratory, Hospital Universitario Galdakao -Usansolo, Galdakao, Vizcaya, Spain.
| |
Collapse
|
2
|
Díez-Campelo M, López-Cadenas F, Xicoy B, Lumbreras E, González T, Del Rey González M, Sánchez-García J, Coll Jordà R, Slama B, Hernández-Rivas JÁ, Thepot S, Bernal T, Guerci-Bresler A, Bargay J, Amigo ML, Preudhomme C, Fenwarth L, Platzbecker U, Götze KS, Arar A, Toribio S, Del Cañizo C, Hernández-Rivas JM, Fenaux P. Low dose lenalidomide versus placebo in non-transfusion dependent patients with low risk, del(5q) myelodysplastic syndromes (SintraREV): a randomised, double-blind, phase 3 trial. Lancet Haematol 2024; 11:e659-e670. [PMID: 39033767 DOI: 10.1016/s2352-3026(24)00142-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 05/02/2024] [Accepted: 05/05/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Lenalidomide is the standard of care for patients who are transfusion dependent with chromosome 5q deletion (del[5q]) myelodysplastic syndromes. In the SintraREV trial, we aimed to investigate whether an early intervention of low lenalidomide doses for 2 years could delay transfusion dependency in patients with anaemia who were not transfusion dependent. METHODS This randomised, double-blind, phase 3 trial, was conducted at 22 sites (University Hospitals) in Spain, France, and Germany. Eligible patients were aged 18 years or older diagnosed with low-risk or intermediate-1-risk del(5q) myelodysplastic syndromes with non-transfusion-dependent anaemia (according to the IPSS), were erythropoietin-stimulating agents naive, and had an ECOG performance status of 2 or less. Patients were randomly assigned (2:1) by means of a telephone system to receive lenalidomide 5 mg daily in 28-day cycles versus placebo for 2 years. The primary endpoint was time to transfusion dependency based on blinded independent central review. Analysis were by intent-to-treat (ITT) and evaluable population. Safety analyses included all participants who received at least one dose of treatment. This trial is registered with ClinicalTrials.gov (NCT01243476) and EudraCT (2009-013619-36) and is complete. FINDINGS Between Feb 15, 2010, and Feb 21, 2018, 61 patients were randomly assigned to receive lenalidomide (n=40; two did not receive treatment) or placebo (n=21). The median age was 72·2 (IQR 65·4-81·9) years, 50 (82%) patients were female, and 11 (18%) were male. The median follow-up time was 60·6 (IQR 32·1-73·9) months. Regarding primary endpoint, median time to transfusion dependency was not reached (95% CI not applicable) in the lenalidomide group versus 11·6 months (95% CI 0·00-30·11) in the placebo group (p=0·0027). Lenalidomide significantly reduced the risk of transfusion dependency by 69·8% (hazard ratio 0·302, 95% CI 0·132-0·692; p=0·0046). The most frequent treatment-related adverse event was neutropenia, occurring in 24 (63%) of 38 patients in the lenalidomide group (grade 3 and 4 in 17 [45%] patients and one [3%], respectively) and in four (19%) of 21 patients in the placebo group (grade 3 in one [5%] patient). Thrombocytopenia was detected in seven (18%) of 38 patients receiving lenalidomide (grade 3 in two [5%] patients). Regarding the non-haematological toxicity, skin disorders (rash nine [23%] of 38 patients) were the most frequently described toxicities among patients receiving lenalidomide, being grade 3 in one (3%) of 38 patients. 19 serious adverse events were reported in 13 patients, 18 in the lenalidomide group and one in the placebo group, five of which were potentially related to the study drug. No treatment-related deaths were identified. INTERPRETATION An early approach with low doses of lenalidomide across two years delays the time to transfusion dependency and improves the rate and quality of the responses, with a manageable safety profile in patients who are non-transfusion dependent with del(5q) low-risk myelodysplastic syndromes. FUNDING Bristol Myers Squibb.
Collapse
Affiliation(s)
- María Díez-Campelo
- Department of Hematology, Hospital Universitario de Salamanca, Salamanca, Spain; IBSAL, Instituto de investigación Biomédica de Salamanca, Salamanca, Spain.
| | - Félix López-Cadenas
- Department of Hematology, Hospital Universitario de Salamanca, Salamanca, Spain; IBSAL, Instituto de investigación Biomédica de Salamanca, Salamanca, Spain
| | - Blanca Xicoy
- Department of Clinical Hematology, Institut Català d'Oncologia, Hospital Germans Trias i Pujol, Barcelona, Spain; Josep Carreras Leukemia Research Institute, Myeloid Neoplasm Group Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Eva Lumbreras
- IBSAL, Instituto de investigación Biomédica de Salamanca, Salamanca, Spain
| | - Teresa González
- IBSAL, Instituto de investigación Biomédica de Salamanca, Salamanca, Spain
| | | | - Joaquín Sánchez-García
- Department of Hematology, Hospital Universitario Reina Sofía, Córdoba, Spain; Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain; Universidad de Córdoba, Córdoba, Spain
| | - Rosa Coll Jordà
- Department of Hematology, Hospital Josep Trueta, Institut Català d'Oncologia, Girona, Spain
| | - Bohrane Slama
- Department of Clinical Hematology, CH Avignon, Avignon, France
| | - Jose-Ángel Hernández-Rivas
- Department of Hematology, Hospital Universitario Infanta Leonor, Universidad Complutense de Madrid, Madrid, Spain
| | - Sylvain Thepot
- Department of Clinical Hematology, Angers University Hospital, Angers, France
| | - Teresa Bernal
- Department of Hematology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | - Joan Bargay
- Department of Hematology, Hospital Universitario Son Llatzer, Palma de Mallorca, Spain; Institut d'Investigacio Illes Balears (IdISBa), Palma de Mallorca, Spain
| | - María Luz Amigo
- Department of Hematology, Hospital Universitario Jose María Morales Meseguer, Murcia, Spain
| | - Claude Preudhomme
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER), UMR9020-U1277, University of Lille, Lille, France; Laboratory of Hematology, CHU Lille, Lille, France
| | - Laurene Fenwarth
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER), UMR9020-U1277, University of Lille, Lille, France; Laboratory of Hematology, CHU Lille, Lille, France
| | - Uwe Platzbecker
- Department for Hematology, Cell Therapy, Hemostaseology, and Infectious Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Katharina S Götze
- Department of Medicine III, Technical University of Munich, Munich, Germany
| | - Ali Arar
- Department of Hematology, CHR Orleans, Orléans, France
| | - Sofía Toribio
- IBSAL, Instituto de investigación Biomédica de Salamanca, Salamanca, Spain
| | - Consuelo Del Cañizo
- Department of Hematology, Hospital Universitario de Salamanca, Salamanca, Spain; IBSAL, Instituto de investigación Biomédica de Salamanca, Salamanca, Spain
| | - Jesús María Hernández-Rivas
- Department of Hematology, Hospital Universitario de Salamanca, Salamanca, Spain; IBSAL, Instituto de investigación Biomédica de Salamanca, Salamanca, Spain; Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Pierre Fenaux
- Département (DMU) d'hématologie et immunologie, Service d'hématologie Seniors, Hôpital St Louis, Université de Paris, APHP Nord Paris, France
| |
Collapse
|
3
|
Urrechaga E, Fernández M, Aguirre U. Complete Blood Counts and Research Parameters in the Detection of Myelodysplastic Syndromes. Diagnostics (Basel) 2024; 14:1322. [PMID: 39001213 PMCID: PMC11240308 DOI: 10.3390/diagnostics14131322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/06/2024] [Accepted: 06/20/2024] [Indexed: 07/16/2024] Open
Abstract
The diagnosis of Myelodysplastic syndromes (MDS) is frequently challenging, especially in terms of the distinction from the other non-neoplastic causes of cytopenia. Currently, it is based on the presence of peripheral blood cytopenias, peripheral blood and bone marrow dysplasia/blasts, and clonal cytogenetic abnormalities, but MDS diagnostic features are polymorphic and non-specific. We investigated the utility of complete blood count (CBC) and research parameters (RUO) from the analyzer BC 6800 Plus (Mindray Diagnostics) to discriminate MDS-related cytopenia. METHODS 100 samples from healthy individuals were used to establish the values of research parameters in normal subjects. A retrospective study was conducted including 66 patients diagnosed with MDS, 90 cytopenic patients due to other diseases (cancer patients receiving therapy, aplastic anemia, other hematological malignancies) and 50 with macrocytic anemia. The Wilcoxon test was applied to detect statistical differences among the groups of patients, considering p < 0.05 significant. The diagnostic performance of the RUO parameters for discriminating MDS among cytopenias was evaluated using receiver operating characteristic (ROC) curve analysis. Amultivariable logistic regression model was performed to identify the potential predictors for having MDS. The area under curve (AUC) and the Hosmer-Lemeshow test of the model were assessed. The performance of the model was verified in a prospective study including 224 cytopenic patients (validation group). RESULTS In the MDS group, the mean cell volume (MCV), percentage of macrocytic red cells (MAC), red cell distribution width (RDW) and immature platelets fraction (IPF) had increased values compared to the cytopenic and normal patients, while platelets, red and white cell counts, Neu X (related to the cytoplasmic complexity of neutrophils), Neu Y (related to nucleic acid content) and Neu Z (related to cell size) were lower (p < 0.001). Neu X, Neu Y, and Neu Z showed higher AUC for detecting MDS > 0.80; MAC, RDW and IPF AUC > 0.76. The multivariable model demonstrated that Neu X and Neu Y could be used in the recognition of MDS, AUC 0.88. In the validation group, 89.0% of the MDS patients were well classified. CONCLUSION MDS are common malignant disorders with a poor prognosis, and early diagnosis is warranted for optimal benefit from treatment. RUO gain insights to detect dysplasia of MDS and could be used in the differential diagnosis of MDS from cytopenias of other etiologies.
Collapse
Affiliation(s)
| | - Mónica Fernández
- Hematology, Hospital Universitario Araba, 01009 Vitoria-Gasteiz, Spain;
| | - Urko Aguirre
- Research Unit, Osakidetza Basque Health Service, Barrualde-Galdakao Integrated Health Organisation, Galdakao-Usansolo Hospital, 48960 Galdakao, Spain;
- Kronikgune Institute for Health Services Research, 48902 Barakaldo, Spain
| |
Collapse
|
4
|
Bruzzese A, Martino EA, Mendicino F, Lucia E, Olivito V, Capodanno I, Neri A, Morabito F, Vigna E, Gentile M. Myelodysplastic syndromes del(5q): Pathogenesis and its therapeutic implications. Eur J Haematol 2024; 112:860-869. [PMID: 38294126 DOI: 10.1111/ejh.14181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/20/2024] [Accepted: 01/22/2024] [Indexed: 02/01/2024]
Abstract
Myelodysplastic syndromes (MDS) encompass a heterogeneous set of acquired bone marrow neoplastic disorders characterized by ineffective hematopoiesis within one or more bone marrow lineages. Nearly half of MDS patients carry cytogenetic alterations, with del(5q) being the most prevalent. Since its first description, del(5q) was consistently correlated with a typical clinical phenotype marked by anemia, thrombocytosis, and a low risk of evolving into acute leukemia. Presently, the World Health Organization (WHO) classification of myeloid neoplasms recognizes a specific subtype of MDS known as "myelodysplastic neoplasm with low blast and isolated del(5q)" identified by the sole presence of 5q deletion or in combination with one other abnormality excluding -7/del(7q). Several studies have sought to unravel the biological processes triggered by del(5q) in the development of MDS, revealing the involvement of various genes localized in specific regions of chromosome 5 referred to as common deleted regions (CDR). This intricate biological landscape makes the MDS cells with del(5q) exceptionally sensitive to lenalidomide. Several studies have confirmed the efficacy of lenalidomide in this context. Regrettably, the response to lenalidomide is not conclusive, prompting ongoing research into biological mechanisms that drive patients toward leukemia and strategies to circumvent lenalidomide resistance and disease progression.
Collapse
Affiliation(s)
- Antonella Bruzzese
- Department of Onco-hematology, Hematology Unit, A.O. of Cosenza, Cosenza, Italy
| | | | - Francesco Mendicino
- Department of Onco-hematology, Hematology Unit, A.O. of Cosenza, Cosenza, Italy
| | - Eugenio Lucia
- Department of Onco-hematology, Hematology Unit, A.O. of Cosenza, Cosenza, Italy
| | - Virginia Olivito
- Department of Onco-hematology, Hematology Unit, A.O. of Cosenza, Cosenza, Italy
| | | | - Antonino Neri
- Scientific Direction Azienda USL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Fortunato Morabito
- Biotechnology Research Unit, Aprigliano, A.O./ASP of Cosenza, Cosenza, Italy
| | - Ernesto Vigna
- Department of Onco-hematology, Hematology Unit, A.O. of Cosenza, Cosenza, Italy
| | - Massimo Gentile
- Department of Onco-hematology, Hematology Unit, A.O. of Cosenza, Cosenza, Italy
- Department of Pharmacy, Health and Nutritional Science, University of Calabria, Rende, Italy
| |
Collapse
|
5
|
Qimudesiren, Yin W, Wang Y, Qing G, Bao J, Chaomurilige, Chen S, Qian L. Hematopoietic Stem Cell Transplantation in the Management of Myelodysplastic Syndrome: A Retrospective, Current, and Future Perspective. Cell Transplant 2024; 33:9636897241284283. [PMID: 39374074 PMCID: PMC11483824 DOI: 10.1177/09636897241284283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 10/08/2024] Open
Abstract
Myelodysplastic syndrome (MDS) is a clonal disorder that affects hematopoietic stem cells (HSCs), primarily occurring in the elderly population. Lower-risk MDS is characterized by a decrease in blood cells, whereas higher-risk MDS is associated with an increased risk of transformation to acute myeloid leukemia (AML). Currently, the treatment of MDS is still unsatisfactory, although demethylating agents, azacitidine (AZA), and decitabine (Dec) have been successfully used to treat MDS and improve survival rates. However, hematopoietic stem cell transplantation (HSCT) remains the only curative treatment for MDS patients, effectively increasing patient survival and quality of life. Nevertheless, treatment-related toxicity, graft-versus-host disease, infectious complications, and relapse are still major post-transplant issues. In this review, through a retrospective analysis of past and present HSCT for the treatment of MDS, we provide insights for the future.
Collapse
Affiliation(s)
- Qimudesiren
- School of Clinical Medicine, Inner Mongolia Minzu University, Tongliao, China
| | - Wenjie Yin
- Department of Hematology, International Mongolian Hospital of Inner Mongolia, Hohhot, China
| | - Yuhong Wang
- Department of Hematology, International Mongolian Hospital of Inner Mongolia, Hohhot, China
| | - Guo Qing
- Department of Hematology, International Mongolian Hospital of Inner Mongolia, Hohhot, China
| | - Jinhua Bao
- Department of Hematology, International Mongolian Hospital of Inner Mongolia, Hohhot, China
| | - Chaomurilige
- Department of Hematology, International Mongolian Hospital of Inner Mongolia, Hohhot, China
- School of Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Shana Chen
- Department of Hematology, International Mongolian Hospital of Inner Mongolia, Hohhot, China
| | - Liren Qian
- Senior Department of Hematology, The Fifth Medical Center of PLA General Hospital, Beijing, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
6
|
An C, Xue F, Sun L, Han H, Zhang Y, Hu Y. The impact of erythroblast enucleation efficiency on the severity of anemia in patients with myelodysplastic syndrome. Cell Commun Signal 2023; 21:332. [PMID: 37986081 PMCID: PMC10658927 DOI: 10.1186/s12964-023-01353-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/11/2023] [Indexed: 11/22/2023] Open
Abstract
Anemia is the most common manifestation in myelodysplastic syndrome (MDS) patients, but the cause of ineffective hematopoiesis is not fully understood. Enucleation is an important event in the maturation process of erythroblasts. According to a series of morphological phenotypes of the pathological development of MDS erythroblasts, we speculate that there may be enucleation disorders. To verify this hypothesis, we cultured MDS bone marrow CD34+ cells in vitro and induced erythroblast development. The results showed that erythroblast enucleation in MDS was significantly lower than that in the normal group, and the rate of enucleation was positively correlated with hemoglobin concentration. Risk stratification of MDS was performed to further analyze the differences in enucleation among the normal group, low-middle risk group and high-risk group. The results showed that the enucleation rate of the high risk group was higher than that of the low-middle risk group but still lower than that of the normal group. Moreover, the expression of pERK and pAKT in MDS erythroblasts in the high risk group was higher than that in the normal group, while the expression of pERK and pAKT in the low-middle risk group was lower than that in the normal group. Furthermore, the enucleation of MDS was positively correlated with the phosphorylation degree of ERK and AKT. In conclusion, this study reveals that the enucleation of erythroblasts is one of the possible causes of anemia in MDS. Video Abstract.
Collapse
Affiliation(s)
- Chao An
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450014, Henan, China.
| | - Fumin Xue
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Ling Sun
- Department of Hematology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Haiyan Han
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450014, Henan, China
| | - Yali Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450014, Henan, China
| | - Yibo Hu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450014, Henan, China.
| |
Collapse
|
7
|
Bruzzese A, Vigna E, Martino EA, Mendicino F, Lucia E, Olivito V, Bova C, Barbato A, Filippelli G, Capodanno I, Neri A, Morabito F, Gentile M. Myelodysplastic syndromes with ring sideroblasts. Hematol Oncol 2023; 41:612-620. [PMID: 36794650 DOI: 10.1002/hon.3125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/18/2022] [Accepted: 02/06/2023] [Indexed: 02/17/2023]
Abstract
Myelodysplastic syndromes (MDS) are acquired bone marrow malignant disorders characterized by ineffective hematopoiesis, resulting from a complex interaction between genetic and epigenetic mutations, alterations of the marrow microenvironment, and the immune system. In 2001, the World Health Organization (WHO) proposed a classification that integrates morphologic and genetic information, considering the MDS with ring sideroblasts (MDS-RS) as a distinct entity. Considering the strong association between MDS-RS and SF3B1 mutation and its importance in the development of MDS, the last WHO classification replaced the prior entity of MDS-RS with MDS with SF3B1 mutation. Several studies were performed to explore this genotype-phenotype correlation. Mutant SF3B1 protein deregulates the expression of genes implicated in developing hematopoietic stem and progenitor cells. Of paramount importance are PPOX and ABCB7 involved in iron metabolism. Another essential role in hemopoiesis is played by the transforming growth factor-beta (TGF-β) receptor. This gene exerts its effects on SMAD pathways, regulating hematopoiesis through effects on balancing proliferation and apoptosis cell inactivity, differentiation, and migration. Luspatercept (ACE-536) is a soluble fusion protein that inhibits molecules in the TGF-β superfamily. Since its structure resembles the TGF-β family receptor, it catches TGF-β superfamily ligands before binding to the receptor, resulting in reduced activation of SMAD signaling, thus enabling erythroid maturation. Luspatercept was investigated in the phase III trial MEDALIST, showing promising efficacy in treating anemia compared to placebo. Nowadays, further studies are needed to explore the real potential of luspatercept, investigating the biological features likely associated with treatment response, the potential use in combination treatments, and its role in the treatment of naïve MDS.
Collapse
Affiliation(s)
- Antonella Bruzzese
- Hematology Unit, Department of Onco-Hematology, AO of Cosenza, Cosenza, Italy
| | - Ernesto Vigna
- Hematology Unit, Department of Onco-Hematology, AO of Cosenza, Cosenza, Italy
| | | | - Francesco Mendicino
- Hematology Unit, Department of Onco-Hematology, AO of Cosenza, Cosenza, Italy
| | - Eugenio Lucia
- Hematology Unit, Department of Onco-Hematology, AO of Cosenza, Cosenza, Italy
| | - Virginia Olivito
- Hematology Unit, Department of Onco-Hematology, AO of Cosenza, Cosenza, Italy
| | - Carlo Bova
- Internal Medicine Department, AO of Cosenza, Cosenza, Italy
| | | | | | | | - Antonino Neri
- Scientific Direction Azienda USL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Fortunato Morabito
- Biotechnology Research Unit, Aprigliano, AO/ASP of Cosenza, Cosenza, Italy
| | - Massimo Gentile
- Hematology Unit, Department of Onco-Hematology, AO of Cosenza, Cosenza, Italy
- Department of Pharmacy, Health and Nutritional Science, University of Calabria, Rende, Italy
| |
Collapse
|
8
|
Doty RT, Lausted CG, Munday AD, Yang Z, Yan X, Meng C, Tian Q, Abkowitz JL. The transcriptomic landscape of normal and ineffective erythropoiesis at single-cell resolution. Blood Adv 2023; 7:4848-4868. [PMID: 37352261 PMCID: PMC10469080 DOI: 10.1182/bloodadvances.2023010382] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/15/2023] [Accepted: 06/07/2023] [Indexed: 06/25/2023] Open
Abstract
The anemias of myelodysplastic syndrome (MDS) and Diamond Blackfan anemia (DBA) are generally macrocytic and always reflect ineffective erythropoiesis yet result from diverse genetic mutations. To delineate shared mechanisms that lead to cell death, we studied the fate of single erythroid marrow cells from individuals with DBA or MDS-5q. We defined an unhealthy (vs healthy) differentiation trajectory using transcriptional pseudotime and cell surface proteins. The pseudotime trajectories diverge immediately after cells upregulate transferrin receptor (CD71), import iron, and initiate heme synthesis, although cell death occurs much later. Cells destined to die express high levels of heme-responsive genes, including ribosomal protein and globin genes, whereas surviving cells downregulate heme synthesis and upregulate DNA damage response, hypoxia, and HIF1 pathways. Surprisingly, 24% ± 12% of cells from control subjects follow the unhealthy trajectory, implying that heme might serve as a rheostat directing cells to live or die. When heme synthesis was inhibited with succinylacetone, more DBA cells followed the healthy trajectory and survived. We also noted high numbers of messages with retained introns that increased as erythroid cells matured, confirmed the rapid cycling of colony forming unit-erythroid, and demonstrated that cell cycle timing is an invariant property of differentiation stage. Including unspliced RNA in pseudotime determinations allowed us to reliably align independent data sets and accurately query stage-specific transcriptomic changes. MDS-5q (unlike DBA) results from somatic mutation, so many normal (unmutated) erythroid cells persist. By independently tracking erythroid differentiation of cells with and without chromosome 5q deletions, we gained insight into why 5q+ cells cannot expand to prevent anemia.
Collapse
Affiliation(s)
- Raymond T. Doty
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA
| | | | - Adam D. Munday
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA
| | - Zhantao Yang
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA
| | | | | | - Qiang Tian
- Institute for Systems Biology, Seattle, WA
| | - Janis L. Abkowitz
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
9
|
Fuchs O. Targeting cereblon in hematologic malignancies. Blood Rev 2023; 57:100994. [PMID: 35933246 DOI: 10.1016/j.blre.2022.100994] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 01/28/2023]
Abstract
The protein cereblon (CRBN) is a substrate receptor of the cullin 4-really interesting new gene (RING) E3 ubiquitin ligase complex CRL4CRBN. Targeting CRBN mediates selective protein ubiquitination and subsequent degradation via the proteasome. This review describes novel thalidomide analogs, immunomodulatory drugs, also known as CRBN E3 ubiquitin ligase modulators or molecular glues (avadomide, iberdomide, CC-885, CC-90009, BTX-1188, CC-92480, CC-99282, CFT7455, and CC-91633), and CRBN-based proteolysis targeting chimeras (PROTACs) with increased efficacy and potent activity for application in hematologic malignancies. Both types of CRBN-binding drugs, molecular glues, and PROTACs stimulate the interaction between CRBN and its neosubstrates, recruiting target disease-promoting proteins and the E3 ubiquitin ligase CRL4CRBN. Proteins that are traditionally difficult to target (transcription factors and oncoproteins) can be polyubiquitinated and degraded in this way. The competition of CRBN neosubstrates with endogenous CRBN-interacting proteins and the pharmacology and rational combination therapies of and mechanisms of resistance to CRL4CRBN modulators or CRBN-based PROTACs are described.
Collapse
Affiliation(s)
- Ota Fuchs
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 12800 Praha 2, Czech Republic.
| |
Collapse
|
10
|
Cappellini MD, Taher AT, Verma A, Shah F, Hermine O. Erythropoiesis in lower-risk myelodysplastic syndromes and beta-thalassemia. Blood Rev 2022; 59:101039. [PMID: 36577601 DOI: 10.1016/j.blre.2022.101039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
The hematologic disorders myelodysplastic syndromes and beta-thalassemia are characterized by ineffective erythropoiesis and anemia, often managed with regular blood transfusions. Erythropoiesis, the process by which sufficient numbers of functional erythrocytes are produced from hematopoietic stem cells, is highly regulated, and defects can negatively affect the proliferation, differentiation, and survival of erythroid precursors. Treatments that directly target the underlying mechanisms of ineffective erythropoiesis are limited, and management of anemia with regular blood transfusions imposes a significant burden on patients, caregivers, and health care systems. There is therefore a strong unmet need for treatments that can restore effective erythropoiesis. Novel therapies are beginning to address this need by targeting a variety of mechanisms underlying erythropoiesis. Herein, we provide an overview of the role of ineffective erythropoiesis in myelodysplastic syndromes and beta-thalassemia, discuss unmet needs in targeting ineffective erythropoiesis, and describe current management strategies and emerging treatments for these disorders.
Collapse
Affiliation(s)
| | - Ali T Taher
- Department of Internal Medicine, American University of Beirut Medical Center, Halim and Aida Daniel Academic and Clinical Center, Beirut, Lebanon.
| | - Amit Verma
- Albert Einstein College of Medicine, New York, NY, USA.
| | - Farrukh Shah
- Department of Haematology, Whittington Health NHS Trust, London, UK.
| | - Olivier Hermine
- Department of Hematology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, University Paris Cité, Paris, France; INSERM U1163 and CNRS 8254, Imagine Institute, Université Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
11
|
Mathieu M, Friedrich C, Ducrot N, Zannoni J, Sylvie T, Jerraya N, Rousseaux S, Chuffart F, Kosmider O, Karim Z, Park S. Luspatercept (RAP-536) modulates oxidative stress without affecting mutation burden in myelodysplastic syndromes. Ann Hematol 2022; 101:2633-2643. [PMID: 36195681 DOI: 10.1007/s00277-022-04993-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 09/23/2022] [Indexed: 11/27/2022]
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.
Collapse
Affiliation(s)
- Meunier Mathieu
- Department of Hematology, CHU Grenoble Alpes, CS10217, 38043, Grenoble cedex 09, France.
- CNRS UMR 5309, INSERM, U1209, Université Grenoble Alpes, Institute for Advanced Bioscience, 38700, Grenoble, France.
| | - Chloé Friedrich
- Institut Cochin, Department Development, Reproduction and Cancer, 75014, Paris, France
| | - Nicolas Ducrot
- Université de Paris, INSERM, CNRS, Centre de Recherche Sur L'Inflammation (CRI), 75018, Paris, France
| | - Johanna Zannoni
- CNRS UMR 5309, INSERM, U1209, Université Grenoble Alpes, Institute for Advanced Bioscience, 38700, Grenoble, France
| | - Tondeur Sylvie
- Laboratoire de Génétique Des Hémopathies, CHU Grenoble Alpes, Grenoble, France
| | - Nelly Jerraya
- CNRS UMR 5309, INSERM, U1209, Université Grenoble Alpes, Institute for Advanced Bioscience, 38700, Grenoble, France
| | - Sophie Rousseaux
- CNRS UMR 5309, INSERM, U1209, Université Grenoble Alpes, Institute for Advanced Bioscience, 38700, Grenoble, France
| | - Florent Chuffart
- CNRS UMR 5309, INSERM, U1209, Université Grenoble Alpes, Institute for Advanced Bioscience, 38700, Grenoble, France
| | - Olivier Kosmider
- Institut Cochin, Department Development, Reproduction and Cancer, 75014, Paris, France
- Hematology Department, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Université de Paris (APHP-CUP), 75014, Paris, France
| | - Zoubida Karim
- Université de Toulouse, INSERM, CNRS, Institut Toulousain Des Maladies Infectieuses Et Inflammatoires (Infinity), Université Paul Sabatier (UPS), Toulouse, France
| | - Sophie Park
- Department of Hematology, CHU Grenoble Alpes, CS10217, 38043, Grenoble cedex 09, France.
- CNRS UMR 5309, INSERM, U1209, Université Grenoble Alpes, Institute for Advanced Bioscience, 38700, Grenoble, France.
| |
Collapse
|
12
|
Morita Y, Nannya Y, Ichikawa M, Hanamoto H, Shibayama H, Maeda Y, Hata T, Miyamoto T, Kawabata H, Takeuchi K, Tanaka H, Kishimoto J, Miyano S, Matsumura I, Ogawa S, Akashi K, Kanakura Y, Mitani K. ASXL1 mutations with serum EPO levels predict poor response to darbepoetin alfa in lower-risk MDS: W-JHS MDS01 trial. Int J Hematol 2022; 116:659-668. [PMID: 35821550 PMCID: PMC9588475 DOI: 10.1007/s12185-022-03414-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 11/25/2022]
Abstract
Darbepoetin alfa (DA) is used to treat anemia in lower-risk (IPSS low or int-1) myelodysplastic syndromes (MDS). However, whether mutations can predict the effectiveness of DA has not been examined. The present study aimed to determine predictive gene mutations. The primary endpoint was a correlation between the presence of highly frequent (≥ 10%) mutations and hematological improvement-erythroid according to IWG criteria 2006 by DA (240 μg/week) until week 16. The study included 79 patients (age 29–90, median 77.0 years; 52 [65.8%] male). Frequently (≥ 10%) mutated genes were SF3B1 (24 cases, 30.4%), TET2 (20, 25.3%), SRSF2 (10, 12.7%), ASXL1 (9, 11.4%), and DNMT3A (8, 10.1%). Overall response rate to DA was 70.9%. Multivariable analysis including baseline erythropoietin levels and red blood cell transfusion volumes as variables revealed that erythropoietin levels and mutations of ASXL1 gene were significantly associated with worse response (odds ratio 0.146, 95% confidence interval 0.042–0.503; p = 0.0023, odds ratio 0.175, 95% confidence interval 0.033–0.928; p = 0.0406, respectively). This study indicated that anemic patients who have higher erythropoietin levels and harbor ASXL1 gene mutations may respond poorly to DA. Alternative strategies are needed for the treatment of anemia in this population. Trial registration number and date of registration: UMIN000022185 and 09/05/2016.
Collapse
Affiliation(s)
- Yasuyoshi Morita
- Divison of Hematology and Rheumatology, Department of Internal Medicine, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Yasuhito Nannya
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan.,Division of Hematopoietic Disease Control, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Motoshi Ichikawa
- Department of Hematology and Oncology, Dokkyo Medical University, 880, Kitakobayashi, Mibu-machi, Shimotsuga-gun, Tochigi, 321-0293, Japan
| | - Hitoshi Hanamoto
- Department of Hematology, Faculty of Medicine, Nara Hospital Kindai University, Nara, Japan
| | - Hirohiko Shibayama
- Department of Hematology, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Yoshinobu Maeda
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| | - Tomoko Hata
- Department of Hematology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Toshihiro Miyamoto
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Science, Fukuoka, Japan
| | - Hiroshi Kawabata
- Department of Hematology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Kazuto Takeuchi
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Hiroko Tanaka
- Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Junji Kishimoto
- Center for Clinical and Translational Research, Kyushu University Hospital, Fukuoka, Japan
| | - Satoru Miyano
- M&D Data Science Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Itaru Matsumura
- Divison of Hematology and Rheumatology, Department of Internal Medicine, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan.,Institute for the Advanced Study of Human Biology (WPI-ASHBi), Department of Medicine, Kyoto University, Kyoto, Japan.,Center for Hematology and Regenerative Medicine, Karolinska Institute, Stockholm, Sweden
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Science, Fukuoka, Japan
| | - Yuzuru Kanakura
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Sumitomo Hospital, Osaka, Japan
| | - Kinuko Mitani
- Department of Hematology and Oncology, Dokkyo Medical University, 880, Kitakobayashi, Mibu-machi, Shimotsuga-gun, Tochigi, 321-0293, Japan.
| |
Collapse
|
13
|
Jouzier C, Cherait A, Cony-Makhoul P, Hamel JF, Veloso M, Thepot S, Cluzeau T, Stamatoullas A, Garnier A, Guerci-Bresler A, Dimicoli-Salazar S, Pica GM, Cheze S, Santana C, Chermat F, Fenaux P, Park S. Red blood cell transfusion burden in myelodysplastic syndromes (MDS) with ring Sideroblasts (RS): A retrospective multicenter study by the Groupe Francophone des Myélodysplasies (GFM). Transfusion 2022; 62:961-973. [PMID: 35452143 DOI: 10.1111/trf.16884] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 02/27/2022] [Accepted: 02/27/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND MDS-RS patients are characterized by chronic anemia and a low risk of Acute Myeloid Leukemia (AML) progression and they generally become Red Blood Cell (RBC) transfusion dependent (TD). STUDY DESIGN AND METHODS We performed a retrospective "real-life" observational study of 6 months in 100 MDS-RS TD patients, recruited in 12 French centers, to describe transfusion characteristics, and evaluate the frequency and causes of hospitalizations, health costs, and morbidity, associated with transfusion dependency, in a French population of RBC transfusion-dependent MDS-RS patients. RESULTS 79% of the patients had high transfusion burden (HTB) and 21% low transfusion burden (LTB). HTB patients had a longer disease duration (6 vs. 3.7 years, p = 0.0078), more frequent iron chelation (82% vs. 50%, p = 0.0052) and higher serum ferritin (p = 0.03). During the 6-month study period, 22% of the patients required inpatient hospitalization, 36% of them for symptomatic anemia requiring emergency RBC transfusion. The 6-month median transfusion costs, including the cost of the day care facility, transportation to and from the hospital, iron chelation, and lab tests, was 16,188€/patient. DISCUSSION MDS-RS represents the archetypal type of chronically transfused lower-risk MDS. Most of those patients have a high transfusion burden and thus frequently need visits to the hospital's day care facility, and frequent hospitalizations, with an overall high median treatment cost. Those costs should be compared with costs of new treatments potentially able to avoid RBC transfusion dependence and to reduce the complications of chronic anemia in MDS-RS patients.
Collapse
Affiliation(s)
- Claire Jouzier
- Service d'hématologie, CHU Grenoble Alpes, Grenoble, France
| | - Amina Cherait
- Service d'hématologie seniors, Hôpital St Louis, Assistance Publique des Hôpitaux de Paris (APHP), Paris, France
| | | | | | - Melanie Veloso
- Service de Biostatistique, CHU Grenoble Alpes, Grenoble, France
| | - Sylvain Thepot
- Service des Maladies du sang, CHU d'Angers, Angers, France
| | | | | | - Alice Garnier
- Service d'hématologie Clinique, CHU de Nantes, France
| | | | | | - Gian Matteo Pica
- Service d'hématologie Clinique, Centre hospitalier Métropole Savoie, Chambéry, France
| | - Stéphane Cheze
- Service d'hématologie Clinique, CHU Côte de Nacre, Caen, France
| | - Clémence Santana
- Service d'hématologie, Centre Léon Bérard, Lyon et en Rhône-Alpes, France
| | - Fatiha Chermat
- Service d'hématologie séniors, Hôpital St Louis, Assistance Publique des Hôpitaux de Paris (APHP), Paris, France
| | - Pierre Fenaux
- Service d'hématologie séniors, Hôpital St Louis, Assistance Publique des Hôpitaux de Paris (APHP), Université de Paris, France
| | - Sophie Park
- Service d'hématologie, CHU Grenoble Alpes, Université Grenoble Alpes, Institute for Advanced Biosciences, Grenoble, France
| |
Collapse
|
14
|
Ak MA, Sahip B, Geduk A, Ucar MA, Kale H, Hacibekiroglu T, Polat MG, Kalpakci Y, Bolaman AZ, Guvenc B, Ertop S. The Clinical Efficacy of Epoetin Alfa and Darbepoetin Alfa in Patients with Low-Risk or Intermediate-1-Risk Myelodysplastic Syndrome: Retrospective Multi-center Real-Life Study. Indian J Hematol Blood Transfus 2022; 38:299-308. [PMID: 35496974 PMCID: PMC9001769 DOI: 10.1007/s12288-021-01458-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/10/2021] [Indexed: 10/21/2022] Open
Abstract
This study aimed to evaluate the clinical efficacy of epoetin alfa and darbepoetin alfa in patients with myelodysplastic syndromes (MDS) in the real-life setting. A total of 204 patients with low-risk or intermediate-1-risk MDS who received epoetin alfa or darbepoetin alfa were included. Hemoglobin levels and transfusion need were recorded before and during 12-month treatment. Hemoglobin levels were significantly higher at each follow up visit when compared to baseline levels in both epoetin alfa (mean ± SD 8.68 ± 1.0 g/dL at baseline vs. 9.83 ± 1.45, 9.99 ± 1.55, 10.24 ± 1.77 and 10.2 ± 1.5 g/dL, respectively) and darbepoetin alfa (8.83 ± 1.09 g/dL at baseline vs. 9.62 ± 1.37, 9.78 ± 1.49, 9.9 ± 1.39 and 10.1 ± 1.5 g/dL, respectively) groups (p < 0.001 for each). Transfusion need significantly decreased from baseline at each study visit in the epoetin alfa group (p < 0.001) and only at the 12th month visit (p < 0.001) in the darbepoetin alfa group. Hemoglobin levels or transfusion need was similar between treatment groups. Overall, 12-month response rate was 58.1% for epoetin alfa and 41.9% for darbepoetin alfa, with no significant difference between treatment groups, whereas higher response rate was noted within the first three months (62.7%) compared to next 9 months (ranged 44.4-60%) of treatment in the epoetin alfa group (p ranged 0.002 to < 0.001). This real-life retrospective study revealed similar efficacy of epoetin alfa and darbepoetin alfa among low risk or intermediate-1 risk MDS patients with no difference in treatment response between treatment groups, whereas a likelihood of earlier treatment response in the epoetin alfa group. Supplementary Information The online version contains supplementary material available at 10.1007/s12288-021-01458-1.
Collapse
Affiliation(s)
- Muzeyyen Aslaner Ak
- Department of Hematology, Zonguldak Bulent Ecevit University Faculty of Medicine, Incivez, 67100 Zonguldak, Turkey
| | - Birsen Sahip
- Department of Hematology, Zonguldak Bulent Ecevit University Faculty of Medicine, Incivez, 67100 Zonguldak, Turkey
| | - Ayfer Geduk
- Department of Hematology, Kocaeli University Faculty of Medicine, Kocaeli, Turkey
| | - Mehmer Ali Ucar
- Department of Hematology, Cukurova University Faculty of Medicine, Adana, Turkey
| | - Hacer Kale
- Department of Hematology, Adnan Menderes University Faculty of Medicine, Aydin, Turkey
| | - Tugba Hacibekiroglu
- Department of Hematology, Sakarya Training and Research Hospital, Sakarya, Turkey
| | - Merve Gokcen Polat
- Department of Hematology, Kocaeli University Faculty of Medicine, Kocaeli, Turkey
| | - Yasin Kalpakci
- Department of Hematology, Sakarya Training and Research Hospital, Sakarya, Turkey
| | - Ali Zahit Bolaman
- Department of Hematology, Adnan Menderes University Faculty of Medicine, Aydin, Turkey
| | - Birol Guvenc
- Department of Hematology, Cukurova University Faculty of Medicine, Adana, Turkey
| | - Sehmus Ertop
- Department of Hematology, Zonguldak Bulent Ecevit University Faculty of Medicine, Incivez, 67100 Zonguldak, Turkey
| |
Collapse
|
15
|
Patnaik MM, Santini V. Targeting ineffective hematopoiesis in myelodysplastic syndromes. Am J Hematol 2022; 97:171-173. [PMID: 34800318 DOI: 10.1002/ajh.26416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 11/17/2021] [Indexed: 12/17/2022]
Affiliation(s)
- Mrinal M. Patnaik
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester Minnesota USA
| | | |
Collapse
|
16
|
Wouters HJCM, Conrads-Frank A, Koinig KA, Smith A, Yu G, de Witte T, Wolffenbuttel BHR, Huls G, Siebert U, Stauder R, van der Klauw MM. The anemia-independent impact of myelodysplastic syndromes on health-related quality of life. Ann Hematol 2021; 100:2921-2932. [PMID: 34476573 PMCID: PMC8592948 DOI: 10.1007/s00277-021-04654-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 08/24/2021] [Indexed: 01/12/2023]
Abstract
Myelodysplastic syndromes (MDS) are in the majority of cases characterized by anemia. Both anemia and MDS per se may directly contribute to impairments in health-related quality of life (HRQoL). In this study, we aimed to investigate the anemia-independent impact of MDS on HRQoL. We evaluated participants (≥ 50 years) from the large population-based Lifelines cohort (N = 44,694, mean age 59.0 ± 7.4 years, 43.6% male) and the European MDS Registry (EUMDS) (N = 1538, mean age 73.4 ± 9.0 years, 63.0% male), which comprises a cohort of lower-risk MDS patients. To enable comparison concerning HRQoL, SF-36 scores measured in Lifelines were converted to EQ-5D-3L index (range 0–1) and dimension scores. Lower-risk MDS patients had significantly lower HRQoL than those from the Lifelines cohort, as illustrated in both the index score and in the five different dimensions. Multivariable linear regression analysis demonstrated that MDS had an adjusted total impact on the EQ-5D index score (B = − 0.12, p < 0.001) and an anemia-independent “direct” impact (B = − 0.10, p < 0.001). Multivariable logistic regression analysis revealed an anemia-independent impact of MDS in the dimension mobility, self-care, usual activities, and anxiety/depression (all except pain/discomfort). This study demonstrates that the major part of the negative impact of lower-risk MDS on HRQoL is not mediated via anemia. Thus, the therapeutic focus should include treatment strategies directed at underlying pathogenic mechanisms to improve HRQoL, rather than aiming predominantly at increasing hemoglobin levels.
Collapse
Affiliation(s)
- Hanneke J C M Wouters
- Department of Hematology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, 9700 RB, Groningen, The Netherlands.
| | - Annette Conrads-Frank
- Institute of Public Health, Medical Decision Making and Health Technology Assessment, Department of Public Health, Health Services Research and Health Technology Assessment, UMIT - University for Health Sciences, Medical Informatics and Technology, Hall in Tirol, Austria
| | - Karin A Koinig
- Department of Internal Medicine V (Hematology and Oncology), Medical University Innsbruck, Innsbruck, Austria
| | - Alex Smith
- Epidemiology and Cancer Statistics Group, Department of Health Sciences, University of York, York, UK
| | - Ge Yu
- Epidemiology and Cancer Statistics Group, Department of Health Sciences, University of York, York, UK
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Theo de Witte
- Department of Tumor Immunology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bruce H R Wolffenbuttel
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, 9700 RB, Groningen, The Netherlands
| | - Gerwin Huls
- Department of Hematology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Uwe Siebert
- Institute of Public Health, Medical Decision Making and Health Technology Assessment, Department of Public Health, Health Services Research and Health Technology Assessment, UMIT - University for Health Sciences, Medical Informatics and Technology, Hall in Tirol, Austria
- Division of Health Technology Assessment, ONCOTYROL - Center for Personalized Cancer Medicine, Innsbruck, Austria
- Center for Health Decision Science, Department of Health Policy and Management, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Institute for Technology Assessment and Department of Radiology and Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Reinhard Stauder
- Department of Internal Medicine V (Hematology and Oncology), Medical University Innsbruck, Innsbruck, Austria
| | - Melanie M van der Klauw
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, 9700 RB, Groningen, The Netherlands
| |
Collapse
|
17
|
Rosenberg CA, Bill M, Rodrigues MA, Hauerslev M, Kerndrup GB, Hokland P, Ludvigsen M. Exploring dyserythropoiesis in patients with myelodysplastic syndrome by imaging flow cytometry and machine-learning assisted morphometrics. CYTOMETRY PART B-CLINICAL CYTOMETRY 2020; 100:554-567. [PMID: 33285035 DOI: 10.1002/cyto.b.21975] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/19/2020] [Accepted: 11/19/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND The hallmark of myelodysplastic syndrome (MDS) remains dysplasia in the bone marrow (BM). However, diagnosing MDS may be challenging and subject to inter-observer variability. Thus, there is an unmet need for novel objective, standardized and reproducible methods for evaluating dysplasia. Imaging flow cytometry (IFC) offers combined analyses of phenotypic and image-based morphometric parameters, for example, cell size and nuclearity. Hence, we hypothesized IFC to be a useful tool in MDS diagnostics. METHODS Using a different-from-normal approach, we investigated dyserythropoiesis by quantifying morphometric features in a median of 5953 erythroblasts (range: 489-68,503) from 14 MDS patients, 11 healthy donors, 6 non-MDS controls with increased erythropoiesis, and 6 patients with cytopenia. RESULTS First, we morphometrically confirmed normal erythroid maturation, as immunophenotypically defined erythroid precursors could be sequenced by significantly decreasing cell-, nuclear- and cytoplasm area. In MDS samples, we demonstrated cell size enlargement and increased fractions of macronormoblasts in late-stage erythroblasts (both p < .0001). Interestingly, cytopenic controls with high-risk mutational patterns displayed highly aberrant cell size morphometrics. Furthermore, assisted by machine learning algorithms, we reliably identified and enumerated true binucleated erythroblasts at a significantly higher frequency in two out of three erythroblast maturation stages in MDS patients compared to normal BM (both p = .0001). CONCLUSION We demonstrate proof-of-concept results of the applicability of automated IFC-based techniques to study and quantify morphometric changes in dyserythropoietic BM cells. We propose that IFC holds great promise as a powerful and objective tool in the complex setting of MDS diagnostics with the potential for minimizing inter-observer variability.
Collapse
Affiliation(s)
| | - Marie Bill
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Mathias Hauerslev
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Gitte B Kerndrup
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Peter Hokland
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Maja Ludvigsen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
18
|
Pilo F, Caocci G, Mele G, La Nasa G. Perls Stain Grade in Bone Marrow Aspirate Correlates with Overall Survival in Low-Risk Myelodysplastic Patients. Acta Haematol 2020; 144:332-336. [PMID: 33011720 DOI: 10.1159/000510111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/13/2020] [Indexed: 01/18/2023]
Abstract
Low-risk patients with myelodysplastic syndromes (MDS) are inclined to long-term accumulation of iron in the organs due mostly to red blood cell transfusion and ineffective erythropoiesis. The effect of free toxic iron species in the liver and heart sites is well known, but recent knowledge assumes that oxidant-mediated tissue injury is also effective in the bone marrow. We aimed to investigate the predictive value of bone marrow iron accumulation as demonstrated by Perls staining on the overall survival (OS) of MDS patients. We retrospectively analyzed 114 low and intermediate-I IPSS risk MDS patients who were diagnosed at our institution in the last 20 years. The median age was 70 years (range 32-93). Two different experienced hematologists analyzed all samples. Perls Prussian blue stain was used to stain the bone marrow, which was assessed by modified Gale's grading and then correlated with the outcome. Twenty-seven patients had grade 1 (+), 31 grade 2 (++), and 56 grade 3 (+++). The 20-year OS was significantly lower in patients with a higher Perls score (6.8 ± 6.1%, median 80 ± 7 months in grade 3; 18.7 ± 9.4%, median 70 ± 17 months in grade 2; 33.2 ± 16.4%, median 144 ± 18 months in grade 1; p = 0.011); bone marrow iron overload (p = 0.003; HR 1.7) and transfusion dependency (0.001; HR 2.6) negatively impacted on survival. We suggest that a higher grade of iron storage at diagnosis can impact on the outcome in MDS patients. Perls stain, together with ferritin and the blood transfusional burden, could be another marker at diagnosis of iron-related toxicity that predicts survival.
Collapse
Affiliation(s)
- Federica Pilo
- Hematology and Bone Marrow Transplant Unit, A. Businco Hospital, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Giovanni Caocci
- Hematology and Bone Marrow Transplant Unit, A. Businco Hospital, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy,
| | - Giannalisa Mele
- Hematology and Bone Marrow Transplant Unit, A. Businco Hospital, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Giorgio La Nasa
- Hematology and Bone Marrow Transplant Unit, A. Businco Hospital, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
19
|
Germing U, Oliva EN, Hiwase D, Almeida A. Treatment of Anemia in Transfusion-Dependent and Non-Transfusion-Dependent Lower-Risk MDS: Current and Emerging Strategies. Hemasphere 2019; 3:e314. [PMID: 31976486 PMCID: PMC6924547 DOI: 10.1097/hs9.0000000000000314] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 10/10/2019] [Accepted: 10/21/2019] [Indexed: 12/17/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of bone marrow disorders with a highly diverse clinical course. For lower-risk MDS patients, therapeutic objectives aim to correct chronic anemia and improve/maintain health-related quality of life (HRQoL). However, disease burden is often insufficiently recognized, and although some patients do not respond/lose response to standard treatment, many are treated late. This is the case for non-transfusion-dependent patients with symptomatic anemia, in whom delayed treatment initiation may lead to unnecessary morbidity. Current active treatment options for lower-risk MDS are limited. Standard care for lower-risk 5q deletion [del(5q)] MDS patients with anemia remains supportive, consisting of red blood cell (RBC) transfusions, iron chelation therapy, and treatment with erythropoiesis-stimulating agents (ESAs) in the case of low serum erythropoietin levels. Response rates to ESAs range from 15% to 63%, whereas 56% to 67% of patients with del(5q) MDS achieve RBC transfusion independence with lenalidomide. Treatment options for patients’ refractory to ESAs and/or lenalidomide, however, are limited. Frequent transfusions are associated with profound clinical, HRQoL, and economic consequences for transfusion-dependent patients. This review focuses on the multiple unmet clinical needs that exist in the treatment of anemia associated with lower-risk MDS and the current and future treatment options that may improve disease management and patient outcomes.
Collapse
Affiliation(s)
- Ulrich Germing
- Department of Hematology, Oncology and Clinical Immunology, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| | - Ester N Oliva
- Department of Hematology, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, Reggio Calabria, Italy
| | - Devendra Hiwase
- Hematology, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Antonio Almeida
- Clinical Hematology, Hospital da Luz Lisboa, Lisbon, Portugal
| |
Collapse
|
20
|
Balleari E, Filiberti RA, Salvetti C, Allione B, Angelucci E, Bruzzone M, Calzamiglia T, Cavaliere M, Cavalleri M, Cilloni D, Clavio M, Crisà E, Da Col A, Danise P, Pilo F, Ferrero D, Finelli C, Gioia D, Lemoli RM, Masiera E, Messa E, Miglino M, Musto P, Natalie Oliva E, Poloni A, Salvi F, Sanna A, Scudeletti M, Tassara R, Santini V. Effects of different doses of erythropoietin in patients with myelodysplastic syndromes: A propensity score-matched analysis. Cancer Med 2019; 8:7567-7576. [PMID: 31657156 PMCID: PMC6912022 DOI: 10.1002/cam4.2638] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 09/15/2019] [Accepted: 10/05/2019] [Indexed: 11/29/2022] Open
Abstract
Background Erythropoiesis‐stimulating agents effectively improve the hemoglobin levels in a fraction of anemic patients with myelodysplastic syndromes (MDS). Higher doses (HD) of recombinant human erythropoietin (rhEPO) have been proposed to overcome suboptimal response rates observed in MDS patients treated with lower “standard doses” (SD) of rhEPO. However, a direct comparison between the different doses of rhEPO is lacking. Methods A cohort of 104 MDS patients treated with HD was retrospectively compared to 208 patients treated with SD in a propensity score‐matched analysis to evaluate hematological improvement‐erythroid (HI‐E) rate induced by the different doses of rhEPO. The impact of rhEPO doses on survival and progression to leukemia was also investigated. Results Overall HI‐E rate was 52.6%. No difference was observed between different rhEPO doses (P = .28) in matched cohorts; in a subgroup analysis, transfusion‐dependent patients and patients with higher IPSS‐R score obtained a higher HI‐E rate with HD, although without significant impact on overall survival (OS). Achievement of HI‐E resulted in superior OS. At univariate analysis, a higher HI‐E rate was observed in transfusion‐independent patients (P < .001), with a lower IPSS‐R score (P < .001) and lower serum EPO levels (P = .027). Multivariate analysis confirmed that rhEPO doses were not significantly related to HI‐E (P = .26). There was no significant difference in OS or progression to leukemia in patients treated with HD vs SD. Conclusion SD are substantially equally effective to HD to improve anemia and influencing survival in MDS patients stratified according to similar propensity to be exposed to rhEPO treatment.
Collapse
Affiliation(s)
- Enrico Balleari
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Internal Medicine, Ospedale Policlinico San Martino-IRCCS, Genova, Italy
| | | | - Chiara Salvetti
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Hematology, University of Turin, Torino, Italy
| | - Bernardino Allione
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Hematology, Molinette Hospital, Torino, Italy
| | - Emanuele Angelucci
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Hematology, Ospedale Policlinico San Martino-IRCCS, Genova, Italy
| | - Marco Bruzzone
- UO Clinical Epidemiology, Ospedale Policlinico San Martino-IRCCS, Genova, Italy
| | - Tullio Calzamiglia
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Internal Medicine-ASL 1, Sanremo (IM), Italy
| | - Marina Cavaliere
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Internal Medicine-ASL 2, Savona, Italy
| | - Maurizio Cavalleri
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Internal Medicine-ASL 4, Sestri Levante (GE), Italy
| | - Daniela Cilloni
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Department of Clinical and Biological Sciences, University of Turin, Torino, Italy
| | - Marino Clavio
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Clinic of Hematology, Department of Internal Medicine, University of Genoa, Ospedale Policlinico San Martino-IRCCS, Genova, Italy
| | - Elena Crisà
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Hematology, University of Turin, Torino, Italy
| | - Anna Da Col
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Internal Medicine, Ospedale Policlinico San Martino-IRCCS, Genova, Italy
| | - Paolo Danise
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Hematology, Nocera Hospital, Nocera Inferiore, Italy
| | - Federica Pilo
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Hematology-P.O. Oncologico Businco AOG. Brotzu, Cagliari, Italy
| | - Dario Ferrero
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Hematology, University of Turin, Torino, Italy
| | - Carlo Finelli
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Hematology, AOU Policlinico Sant'Orsola-Malpighi, University of Bologna, Bologna, Italy
| | - Daniela Gioia
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy
| | - Roberto Massimo Lemoli
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Clinic of Hematology, Department of Internal Medicine, University of Genoa, Ospedale Policlinico San Martino-IRCCS, Genova, Italy
| | - Elisa Masiera
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy
| | - Emanuela Messa
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Internal Medicine, ASLTo4, Carmagnola, Italy
| | - Maurizio Miglino
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Clinic of Hematology, Department of Internal Medicine, University of Genoa, Ospedale Policlinico San Martino-IRCCS, Genova, Italy
| | - Pellegrino Musto
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,Regional Department of Hematology, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture (Pz), Italy
| | - Esther Natalie Oliva
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Hematology, Grande Ospedale Metropolitano "Bianchi Melacrino Morelli", Reggio Calabria, Italy
| | - Antonella Poloni
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Hematology, Università Politecnica delle Marche, Ancona, Italy
| | - Flavia Salvi
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Hematology, SS. Antonio e Biagio Hospital, Alessandria, Italy
| | - Alessandro Sanna
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,Ematologia, Ospedale di Livorno, Livorno, Italy
| | - Marco Scudeletti
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Internal Medicine-ASL 4, Sestri Levante (GE), Italy
| | - Rodolfo Tassara
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,UO Internal Medicine-ASL 2, Savona, Italy
| | - Valeria Santini
- Fondazione Italiana Sindromi Mielodisplastiche (FISM), Bologna, Italy.,MDS Unit, AOU Careggi, University of Florence, Firenze, Italy
| |
Collapse
|
21
|
Moura ATG, Duarte FB, Barbosa MC, de Jesus dos Santos TE, Lemes RPG. Prolonged response to recombinant human erythropoietin treatment in patients with myelodysplastic syndrome at a single referral centre in Brazil. Clinics (Sao Paulo) 2019; 74:e771. [PMID: 31508719 PMCID: PMC6724462 DOI: 10.6061/clinics/2019/e771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 05/30/2019] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES To evaluate the effects of epoetin (EPO) alfa treatment on overall survival, event-free survival and response duration in patients with myelodysplastic syndrome (MDS) who were treated at a haematological referral centre in northeastern Brazil. METHODS This was a retrospective cohort study of 36 patients diagnosed with MDS and treated with EPO alfa at 30,000 to 60,000 IU per week. Clinical data were collected from medical records. The events assessed were non-response to treatment and progression to acute myeloid leukaemia (AML). Statistical analyses were performed using GraphPad Prism 7 and SPSS 24 software. RESULTS The overall survival of patients who received EPO alfa treatment was 51.64%, with a median of 65 months of treatment, and the overall survival of this group was 100% during the first 24 months. We detected a 43.5-month median event-free survival, with a response rate of 80.5%. We observed responses from 25 to 175 months. Patients with transfusion dependence and those with a high-risk stratification, as determined by the International Prognostic Scoring System (IPSS), the Revised International Prognostic Scoring System (IPSS-R), the WHO classification-based Prognostic Scoring System (WPSS) and the WHO 2016, had a lower event-free survival than other patients. CONCLUSIONS Despite the wide use of EPO alfa in the treatment of anaemia in patients with MDS, the median response duration is approximately only 24 months. Our data provide encouraging results concerning the benefits of using EPO alfa for the improvement of the quality of life, as patients treated with EPO showed higher overall survival, event-free survival rates and longer response durations than have been previously described in the literature.
Collapse
Affiliation(s)
- Anna Thawanny Gadelha Moura
- Laboratorio de Pesquisa em Hemoglobinopatias e Genetica das Doencas Hematologicas, Universidade Federal do Ceara, Fortaleza, CE, BR
| | | | - Maritza Cavalcante Barbosa
- Laboratorio de Pesquisa em Hemoglobinopatias e Genetica das Doencas Hematologicas, Universidade Federal do Ceara, Fortaleza, CE, BR
| | - Talyta Ellen de Jesus dos Santos
- Laboratorio de Pesquisa em Hemoglobinopatias e Genetica das Doencas Hematologicas, Universidade Federal do Ceara, Fortaleza, CE, BR
| | - Romélia Pinheiro Gonçalves Lemes
- Laboratorio de Pesquisa em Hemoglobinopatias e Genetica das Doencas Hematologicas, Universidade Federal do Ceara, Fortaleza, CE, BR
| |
Collapse
|
22
|
Ye F, Li N. Role of p15(INK4B) Methylation in Patients With Myelodysplastic Syndromes: A Systematic Meta-Analysis. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 19:e259-e265. [PMID: 31023595 DOI: 10.1016/j.clml.2019.03.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 03/17/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Tumor suppressor gene cyclin-dependent kinase inhibitor 2B (p15(INK4B)) methylation has been frequently reported in myelodysplastic syndromes (MDS). However, the association between p15(INK4B) methylation and MDS remains elusive. Thus, this meta-analysis was first conducted to evaluate the clinical significance of p15(INK4B) methylation in MDS. MATERIALS AND METHODS Eligible studies were identified via an online electronic databases search. The overall odds ratios (ORs) or hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated. RESULTS Twenty-eight studies published between 1997 and 2017 were identified, including 1205 MDS patients and 243 nontumor controls. No evidence of heterogeneity was found in our study. p15(INK4B) methylation was significantly elevated in MDS compared with nontumor controls (OR, 10.37; P < .001). In addition, p15(INK4B) methylation was significantly higher in advanced MDS than in early MDS (OR, 4.70; P < .001) and was linked to an unfavorable overall survival (multivariate analysis: HR, 1.78; 95% CI, 1.23-2.71). Subgroup analyses on the basis of ethnicity and detection method showed that the results remained significant in different subgroups (all Ps < .05). CONCLUSION Our findings suggest that p15(INK4B) methylation might play an important role in the development, progression, and poor prognosis of MDS. More prospective studies with larger study populations are needed.
Collapse
Affiliation(s)
- Fang Ye
- Department of Hematology, Chuiyangliu Hospital affiliated to Tsinghua University, Beijing, China.
| | - Ningning Li
- Department of Hematology, Chuiyangliu Hospital affiliated to Tsinghua University, Beijing, China
| |
Collapse
|
23
|
Fuchs O. Treatment of Lymphoid and Myeloid Malignancies by Immunomodulatory Drugs. Cardiovasc Hematol Disord Drug Targets 2019; 19:51-78. [PMID: 29788898 DOI: 10.2174/1871529x18666180522073855] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 05/05/2018] [Accepted: 05/14/2018] [Indexed: 06/08/2023]
Abstract
Thalidomide and its derivatives (lenalidomide, pomalidomide, avadomide, iberdomide hydrochoride, CC-885 and CC-90009) form the family of immunomodulatory drugs (IMiDs). Lenalidomide (CC5013, Revlimid®) was approved by the US FDA and the EMA for the treatment of multiple myeloma (MM) patients, low or intermediate-1 risk transfusion-dependent myelodysplastic syndrome (MDS) with chromosome 5q deletion [del(5q)] and relapsed and/or refractory mantle cell lymphoma following bortezomib. Lenalidomide has also been studied in clinical trials and has shown promising activity in chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma (NHL). Lenalidomide has anti-inflammatory effects and inhibits angiogenesis. Pomalidomide (CC4047, Imnovid® [EU], Pomalyst® [USA]) was approved for advanced MM insensitive to bortezomib and lenalidomide. Other IMiDs are in phases 1 and 2 of clinical trials. Cereblon (CRBN) seems to have an important role in IMiDs action in both lymphoid and myeloid hematological malignancies. Cereblon acts as the substrate receptor of a cullin-4 really interesting new gene (RING) E3 ubiquitin ligase CRL4CRBN. This E3 ubiquitin ligase in the absence of lenalidomide ubiquitinates CRBN itself and the other components of CRL4CRBN complex. Presence of lenalidomide changes specificity of CRL4CRBN which ubiquitinates two transcription factors, IKZF1 (Ikaros) and IKZF3 (Aiolos), and casein kinase 1α (CK1α) and marks them for degradation in proteasomes. Both these transcription factors (IKZF1 and IKZF3) stimulate proliferation of MM cells and inhibit T cells. Low CRBN level was connected with insensitivity of MM cells to lenalidomide. Lenalidomide decreases expression of protein argonaute-2, which binds to cereblon. Argonaute-2 seems to be an important drug target against IMiDs resistance in MM cells. Lenalidomide decreases also basigin and monocarboxylate transporter 1 in MM cells. MM cells with low expression of Ikaros, Aiolos and basigin are more sensitive to lenalidomide treatment. The CK1α gene (CSNK1A1) is located on 5q32 in commonly deleted region (CDR) in del(5q) MDS. Inhibition of CK1α sensitizes del(5q) MDS cells to lenalidomide. CK1α mediates also survival of malignant plasma cells in MM. Though, inhibition of CK1α is a potential novel therapy not only in del(5q) MDS but also in MM. High level of full length CRBN mRNA in mononuclear cells of bone marrow and of peripheral blood seems to be necessary for successful therapy of del(5q) MDS with lenalidomide. While transfusion independence (TI) after lenalidomide treatment is more than 60% in MDS patients with del(5q), only 25% TI and substantially shorter duration of response with occurrence of neutropenia and thrombocytopenia were achieved in lower risk MDS patients with normal karyotype treated with lenalidomide. Shortage of the biomarkers for lenalidomide response in these MDS patients is the main problem up to now.
Collapse
Affiliation(s)
- Ota Fuchs
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic
| |
Collapse
|
24
|
Ru YX, Dong SX, Li Y, Zhao SX, Liang HY, Zhu XF, Zheng YZ, Zhang FK. A novel anemia associated with membranous cytoplasm degeneration in 16 patients: an ultrastructural study. Ultrastruct Pathol 2018; 42:350-357. [PMID: 29913101 DOI: 10.1080/01913123.2018.1485807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Sixteen patients with mild anemia and hemolysis were difficult to be classified into any known category based on laboratory examinations and light microscopy. To make a definite diagnosis and investigate the pathomechanism, ultrastructural study was performed on erythroid cells from 16 patients. Transmission electron microscopy demonstrated a series of alterations of cytoplasm, including cytoplasm sequestration, membranous transformation, and degeneration in erythroblasts and reticulocytes at different stages. The affected erythroblasts were usually complicated with chromatin condensation, karyorrhexis, nuclear membrane lysis, and megaloblastic changes. The reticulocytes with the cytoplasm alterations had a huge size from 10 um to 15 um in diameter. The membranous cytoplasm degeneration revealed a unique pathomechanism of dyserythropoiesis and ineffective erythropoiesis in 16 patients with anemia, and suggested a novel anemia category though more details remained to be investigated.
Collapse
Affiliation(s)
- Yong-Xin Ru
- a State Key Laboratory of Experimental Hematology, Peking Union Medical College , Institute of Hematology & Blood Diseases Hospital , Beijing , China
| | - Shu-Xu Dong
- a State Key Laboratory of Experimental Hematology, Peking Union Medical College , Institute of Hematology & Blood Diseases Hospital , Beijing , China
| | - Yuan Li
- a State Key Laboratory of Experimental Hematology, Peking Union Medical College , Institute of Hematology & Blood Diseases Hospital , Beijing , China
| | - Shi-Xuan Zhao
- a State Key Laboratory of Experimental Hematology, Peking Union Medical College , Institute of Hematology & Blood Diseases Hospital , Beijing , China
| | - Hao-Yue Liang
- a State Key Laboratory of Experimental Hematology, Peking Union Medical College , Institute of Hematology & Blood Diseases Hospital , Beijing , China
| | - Xiao-Fan Zhu
- a State Key Laboratory of Experimental Hematology, Peking Union Medical College , Institute of Hematology & Blood Diseases Hospital , Beijing , China
| | - Yi-Zhou Zheng
- a State Key Laboratory of Experimental Hematology, Peking Union Medical College , Institute of Hematology & Blood Diseases Hospital , Beijing , China
| | - Feng-Kui Zhang
- a State Key Laboratory of Experimental Hematology, Peking Union Medical College , Institute of Hematology & Blood Diseases Hospital , Beijing , China
| |
Collapse
|
25
|
Santini V, Almeida A, Giagounidis A, Platzbecker U, Buckstein R, Beach C, Guo S, Altincatal A, Wu C, Fenaux P. The Effect of Lenalidomide on Health-Related Quality of Life in Patients With Lower-Risk Non-del(5q) Myelodysplastic Syndromes: Results From the MDS-005 Study. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2018; 18:136-144.e7. [DOI: 10.1016/j.clml.2017.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/26/2017] [Indexed: 11/24/2022]
|
26
|
Komrokji R, Swern AS, Grinblatt D, Lyons RM, Tobiasson M, Silverman LR, Sayar H, Vij R, Fliss A, Tu N, Sugrue MM. Azacitidine in Lower-Risk Myelodysplastic Syndromes: A Meta-Analysis of Data from Prospective Studies. Oncologist 2018; 23:159-170. [PMID: 29118268 PMCID: PMC5813747 DOI: 10.1634/theoncologist.2017-0215] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 09/06/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND After erythropoiesis-stimulating agent (ESA) failure, lenalidomide and hypomethylating agents are the only remaining treatment options for most patients with lower-risk myelodysplastic syndromes (LR-MDS). Optimal choice of these agents as front-line therapy in non-del(5q) LR-MDS is unclear. Because azacitidine clinical data mainly describe experience in higher-risk MDS, we performed a meta-analysis of patient-level data to evaluate azacitidine in patients with red blood cell (RBC) transfusion-dependent LR-MDS. MATERIALS AND METHODS We searched English-language articles for prospective phase II and III azacitidine clinical trials and patient registries published between 2000 and 2015, and Embase abstracts from 2015 conferences. Patient-level data from identified relevant studies were provided by investigators. Meta-analyses followed Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Efficacy endpoints were RBC transfusion independence (TI) and Clinical Benefit (RBC-TI, erythroid response, and complete or partial remission, per International Working Group 2006 criteria for MDS). RESULTS Data for 233 patients from 6 clinical studies and 1 registry study met criteria for inclusion in analyses. Overall, 90.3% of patients had non-del(5q) LR-MDS. Pooled estimates from random-effects models of RBC-TI and Clinical Benefit were 38.9% and 81.1%, respectively; for the ESA-refractory subgroup, they were 40.5% and 77.3%; and for patients with isolated anemia, they were 41.9% and 82.5%. In multivariate analyses, planned use of ≥6 azacitidine treatment cycles was significantly predictive of response. CONCLUSION Azacitidine effects in these patients, most with non-del(5q) LR-MDS, were promising and generally similar to those reported for lenalidomide in similar patients. The choice of initial therapy is important because most patients eventually stop responding to front-line therapy and alternatives are limited. IMPLICATIONS FOR PRACTICE Lower-risk myelodysplastic syndromes (LR-MDS) are primarily characterized by anemia. After erythropoiesis-stimulating agent (ESA) failure, lenalidomide and hypomethylating agents are the only remaining treatment options for most patients. This meta-analysis of 233 azacitidine-treated red blood cell (RBC) transfusion-dependent patients with LR-MDS (92.3% non-del[5q]) from 7 studies showed 38.9% became RBC transfusion-independent. There is no clear guidance regarding the optimal choice of lenalidomide or hypomethylating agents for patients with non-del(5q) LR-MDS following ESA failure. Clinical presentation (e.g., number of cytopenias) and potential outcomes after hypomethylating agent failure are factors to consider when making initial treatment decisions for LR-MDS patients.
Collapse
Affiliation(s)
- Rami Komrokji
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Arlene S Swern
- Biostatistics, Celgene Corporation, Summit, New Jersey, USA
| | - David Grinblatt
- Hematology, Northshore University Health System, Evanston, Illinois, USA
| | - Roger M Lyons
- Department of Hematology, US Oncology-Texas Oncology, San Antonio, Texas, USA
| | - Magnus Tobiasson
- Division of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Lewis R Silverman
- Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Hamid Sayar
- Simon Cancer Center, Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Ravi Vij
- Department of Internal Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Albert Fliss
- Medical Affairs, Celgene Corporation, Summit, New Jersey, USA
| | - Nora Tu
- Biostatistics, Celgene Corporation, Summit, New Jersey, USA
| | - Mary M Sugrue
- Medical Affairs, Celgene Corporation, Summit, New Jersey, USA
| |
Collapse
|
27
|
Almeida A, Fenaux P, Garcia-Manero G, Goldberg SL, Gröpper S, Jonasova A, Vey N, Castaneda C, Zhong J, Beach CL, Santini V. Safety profile of lenalidomide in patients with lower-risk myelodysplastic syndromes without del(5q): results of a phase 3 trial. Leuk Lymphoma 2018; 59:2135-2143. [PMID: 29322849 DOI: 10.1080/10428194.2017.1421758] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The safety profile of lenalidomide use in lower-risk myelodysplastic syndromes (MDS) patients with del(5q) is well-established, but less is known in non-del(5q) patients. We provide safety data from a randomized, phase 3 trial evaluating lenalidomide in 239 patients with lower-risk non-del(5q) MDS ineligible/refractory to erythropoiesis-stimulating agents (ESAs). Compared with placebo, lenalidomide was associated with a higher incidence of grade 3-4 treatment-emergent adverse events (TEAEs; 86% vs. 44%), but not risk of infection (p = .817) or hemorrhagic events (p = 1.000). Grade 3-4 non-hematologic TEAEs were rare (the incidence of grade 3-4 pneumonia, e.g. was 5.6% in the lenalidomide group and 2.5% in the placebo group). Common grade 1-2 non-hematologic TEAEs did not require dose modifications or treatment discontinuation. Acute myeloid leukemia and second primary malignancies incidence was similar across treatment groups. Lenalidomide had a predictable and manageable safety profile in lower-risk non-del(5q) MDS patients ineligible/refractory to ESAs. Guidance on managing lenalidomide-related TEAEs is provided to help maintain patients on therapy to achieve maximum clinical benefit. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT01029262.
Collapse
Affiliation(s)
- Antonio Almeida
- a Departamento de Hematologia , Instituto Português de Oncologia de Lisboa Francisco Gentil , Lisbon , Portugal
| | - Pierre Fenaux
- b Service d'Hématologie Séniors, Hôpital Saint-Louis, Université Paris 7 , Paris , France
| | | | - Stuart L Goldberg
- d John Theurer Cancer Center, Hackensack University Medical Center , Hackensack , NJ , USA
| | | | - Anna Jonasova
- f First Faculty of Medicine , Charles University General Hospital , Prague , Czech Republic
| | - Norbert Vey
- g Institut Paoli-Calmettes, Centre Régional de Lutte Contre le Cancer , Marseilles , France
| | | | | | - C L Beach
- h Celgene Corporation , Summit , NJ , USA
| | - Valeria Santini
- i Azienda Ospedaliero Universitaria Careggi, University of Florence , Florence , Italy
| |
Collapse
|
28
|
Santini V. Defeating anaemia in myelodysplastic syndromes: another step forward. Lancet Oncol 2017; 18:1290-1292. [DOI: 10.1016/s1470-2045(17)30671-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 08/11/2017] [Indexed: 11/30/2022]
|
29
|
Sanchez-Garcia J, Falantes J, Medina Perez A, Hernandez-Mohedo F, Hermosin L, Torres-Sabariego A, Bailen A, Hernandez-Sanchez JM, Solé Rodriguez M, Casaño FJ, Calderon C, Labrador M, Vahí M, Serrano J, Lumbreras E, Hernández-Rivas JM. Prospective randomized trial of 5 days azacitidine versus supportive care in patients with lower-risk myelodysplastic syndromes without 5q deletion and transfusion-dependent anemia. Leuk Lymphoma 2017; 59:1095-1104. [DOI: 10.1080/10428194.2017.1366998] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Joaquin Sanchez-Garcia
- Hematology Department, Hospital Universitario Reina Sofía, Instituto Maimonides Investigación Biomédica, IMIBIC, University of Córdoba, Córdoba, Spain
| | - Jose Falantes
- Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS), CSIC, Universidad de Sevilla, Sevilla, Spain
| | | | | | | | | | | | - Jesus M. Hernandez-Sanchez
- Hospital Universitario de Salamanca, IBSAL, UDMCC, Centro de Investigación del Cancer (Universidad de Salamanca, CSIC), Salamanca, Spain
| | - María Solé Rodriguez
- Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS), CSIC, Universidad de Sevilla, Sevilla, Spain
| | - Francisco Javier Casaño
- Hematology Department, Hospital Universitario Reina Sofía, Instituto Maimonides Investigación Biomédica, IMIBIC, University of Córdoba, Córdoba, Spain
| | - Cristina Calderon
- Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS), CSIC, Universidad de Sevilla, Sevilla, Spain
| | | | | | - Josefina Serrano
- Hematology Department, Hospital Universitario Reina Sofía, Instituto Maimonides Investigación Biomédica, IMIBIC, University of Córdoba, Córdoba, Spain
| | - Eva Lumbreras
- Hospital Universitario de Salamanca, IBSAL, UDMCC, Centro de Investigación del Cancer (Universidad de Salamanca, CSIC), Salamanca, Spain
| | - Jesus Maria Hernández-Rivas
- Hospital Universitario de Salamanca, IBSAL, UDMCC, Centro de Investigación del Cancer (Universidad de Salamanca, CSIC), Salamanca, Spain
| | | |
Collapse
|
30
|
Oben KZ, Alhakeem SS, McKenna MK, Brandon JA, Mani R, Noothi SK, Jinpeng L, Akunuru S, Dhar SK, Singh IP, Liang Y, Wang C, Abdel-Latif A, Stills HF, St Clair DK, Geiger H, Muthusamy N, Tohyama K, Gupta RC, Bondada S. Oxidative stress-induced JNK/AP-1 signaling is a major pathway involved in selective apoptosis of myelodysplastic syndrome cells by Withaferin-A. Oncotarget 2017; 8:77436-77452. [PMID: 29100399 PMCID: PMC5652791 DOI: 10.18632/oncotarget.20497] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 07/16/2017] [Indexed: 02/07/2023] Open
Abstract
Myelodysplastic syndromes (MDS) are a diverse group of malignant clonal hematopoietic stem cell disorders characterized by ineffective hematopoiesis, dysplastic cell morphology in one or more hematopoietic lineages, and a risk of progression to acute myeloid leukemia (AML). Approximately 50% of MDS patients respond to current FDA-approved drug therapies but a majority of responders relapse within 2-3 years. There is therefore a compelling need to identify potential new therapies for MDS treatment. We utilized the MDS-L cell line to investigate the anticancer potential and mechanisms of action of a plant-derived compound, Withaferin A (WFA), in MDS. WFA was potently cytotoxic to MDS-L cells but had no significant effect on the viability of normal human primary bone marrow cells. WFA also significantly reduced engraftment of MDS-L cells in a xenotransplantation model. Through transcriptome analysis, we identified reactive oxygen species (ROS)-activated JNK/AP-1 signaling as a major pathway mediating apoptosis of MDS-L cells by WFA. We conclude that the molecular mechanism mediating selective cytotoxicity of WFA on MDS-L cells is strongly associated with induction of ROS. Therefore, pharmacologic manipulation of redox biology could be exploited as a selective therapeutic target in MDS.
Collapse
Affiliation(s)
- Karine Z Oben
- Markey Cancer Center and Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| | - Sara S Alhakeem
- Markey Cancer Center and Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| | - Mary K McKenna
- Markey Cancer Center and Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| | - Jason A Brandon
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Rajeswaran Mani
- Comprehensive Cancer Center and Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Sunil K Noothi
- Markey Cancer Center and Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| | - Liu Jinpeng
- Biostatistics Core, Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Shailaja Akunuru
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Sanjit K Dhar
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Inder P Singh
- Department of Natural Products, National Institute of Pharmaceutical Research, S.A.S Nagar, Punjab 160062, India
| | - Ying Liang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Chi Wang
- Biostatistics Core, Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Ahmed Abdel-Latif
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Harold F Stills
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| | - Daret K St Clair
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Hartmut Geiger
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Natarajan Muthusamy
- Comprehensive Cancer Center and Department of Internal Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Kaoru Tohyama
- Department of Laboratory Medicine, Kawasaki Medical School, Kurashiki, Okayama 701-0192, Japan
| | - Ramesh C Gupta
- Department of Pharmacology and Toxicology, and James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Subbarao Bondada
- Markey Cancer Center and Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
31
|
Yip BH, Steeples V, Repapi E, Armstrong RN, Llorian M, Roy S, Shaw J, Dolatshad H, Taylor S, Verma A, Bartenstein M, Vyas P, Cross NC, Malcovati L, Cazzola M, Hellström-Lindberg E, Ogawa S, Smith CW, Pellagatti A, Boultwood J. The U2AF1S34F mutation induces lineage-specific splicing alterations in myelodysplastic syndromes. J Clin Invest 2017; 127:2206-2221. [PMID: 28436936 PMCID: PMC5451246 DOI: 10.1172/jci91363] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 02/21/2017] [Indexed: 12/23/2022] Open
Abstract
Mutations of the splicing factor–encoding gene U2AF1 are frequent in the myelodysplastic syndromes (MDS), a myeloid malignancy, and other cancers. Patients with MDS suffer from peripheral blood cytopenias, including anemia, and an increasing percentage of bone marrow myeloblasts. We studied the impact of the common U2AF1S34F mutation on cellular function and mRNA splicing in the main cell lineages affected in MDS. We demonstrated that U2AF1S34F expression in human hematopoietic progenitors impairs erythroid differentiation and skews granulomonocytic differentiation toward granulocytes. RNA sequencing of erythroid and granulomonocytic colonies revealed that U2AF1S34F induced a higher number of cassette exon splicing events in granulomonocytic cells than in erythroid cells. U2AF1S34F altered mRNA splicing of many transcripts that were expressed in both cell types in a lineage-specific manner. In hematopoietic progenitors, the introduction of isoform changes identified in the U2AF1S34F target genes H2AFY, encoding an H2A histone variant, and STRAP, encoding serine/threonine kinase receptor–associated protein, recapitulated phenotypes associated with U2AF1S34F expression in erythroid and granulomonocytic cells, suggesting a causal link. Furthermore, we showed that isoform modulation of H2AFY and STRAP rescues the erythroid differentiation defect in U2AF1S34F MDS cells, suggesting that splicing modulators could be used therapeutically. These data have critical implications for understanding MDS phenotypic heterogeneity and support the development of therapies targeting splicing abnormalities.
Collapse
Affiliation(s)
- Bon Ham Yip
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and BRC Blood Theme, National Institute for Health Research (NIHR) Oxford Biomedical Centre, Oxford University Hospital, Oxford, United Kingdom
| | - Violetta Steeples
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and BRC Blood Theme, National Institute for Health Research (NIHR) Oxford Biomedical Centre, Oxford University Hospital, Oxford, United Kingdom
| | - Emmanouela Repapi
- The Computational Biology Research Group, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Richard N Armstrong
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and BRC Blood Theme, National Institute for Health Research (NIHR) Oxford Biomedical Centre, Oxford University Hospital, Oxford, United Kingdom
| | - Miriam Llorian
- Department of Biochemistry, Downing Site, University of Cambridge, Cambridge, United Kingdom
| | - Swagata Roy
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and BRC Blood Theme, National Institute for Health Research (NIHR) Oxford Biomedical Centre, Oxford University Hospital, Oxford, United Kingdom
| | - Jacqueline Shaw
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and BRC Blood Theme, National Institute for Health Research (NIHR) Oxford Biomedical Centre, Oxford University Hospital, Oxford, United Kingdom
| | - Hamid Dolatshad
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and BRC Blood Theme, National Institute for Health Research (NIHR) Oxford Biomedical Centre, Oxford University Hospital, Oxford, United Kingdom
| | - Stephen Taylor
- The Computational Biology Research Group, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Amit Verma
- Albert Einstein College of Medicine, Bronx, New York, USA
| | | | - Paresh Vyas
- Medical Research Council, Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, and Department of Hematology, Oxford University Hospital National Health Service Trust, Oxford, United Kingdom
| | - Nicholas Cp Cross
- Faculty of Medicine, University of Southampton, Southampton, and National Genetics Reference Laboratory (Wessex), Salisbury, United Kingdom
| | - Luca Malcovati
- Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Mario Cazzola
- Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Eva Hellström-Lindberg
- Center for Hematology and Regenerative Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | - Christopher Wj Smith
- Department of Biochemistry, Downing Site, University of Cambridge, Cambridge, United Kingdom
| | - Andrea Pellagatti
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and BRC Blood Theme, National Institute for Health Research (NIHR) Oxford Biomedical Centre, Oxford University Hospital, Oxford, United Kingdom
| | - Jacqueline Boultwood
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and BRC Blood Theme, National Institute for Health Research (NIHR) Oxford Biomedical Centre, Oxford University Hospital, Oxford, United Kingdom
| |
Collapse
|
32
|
Xiao ZJ. [How I treat myelodysplastic symdromes]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2017; 38:268-271. [PMID: 28468085 PMCID: PMC7342718 DOI: 10.3760/cma.j.issn.0253-2727.2017.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Indexed: 11/05/2022]
|
33
|
Myelodysplastic Syndrome Clinically Presenting with the "Classic TTP Pentad". Case Rep Hematol 2017; 2017:4619406. [PMID: 28255478 PMCID: PMC5309395 DOI: 10.1155/2017/4619406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 01/06/2017] [Accepted: 01/16/2017] [Indexed: 11/17/2022] Open
Abstract
The clinical presentation of myelodysplastic syndrome (MDS) is not specific. Many patients can be asymptomatic and can be detected only due to an abnormal complete blood cell count (CBC) on routine exam or for other reasons while others can be symptomatic as a consequence of underlying cytopenias. Thrombotic thrombocytopenic purpura (TTP) usually is suspected under the evidence of microangiopathic hemolytic anemia (MAHA) and thrombocytopenia and because it is a life-threatening condition (medical emergency) immediate initiation of plasmapheresis could be life-saving. The following case illustrates an unusual presentation of MDS in a patient who came in to the emergency room with the classic TTP “pentad” of fever, renal involvement, MAHA, mental status changes, and thrombocytopenia. We will focus our discussion in the clinical presentation of this case.
Collapse
|
34
|
Zhang Y, Qian LL, Shen JP, Chen JF, Gao YT, Xiang JJ, Ye BD, Zhou YH. Effect of Chinese medicine treatment based on pattern identification on cellular immunophenotype of myelodysplastic syndrome. Chin J Integr Med 2016; 23:469-473. [PMID: 27933512 DOI: 10.1007/s11655-016-2276-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To observe the influence of treatment based on Chinese medicine pattern identification on cellular immunophenotype of the myelodysplastic syndrome (MDS). METHODS Sixty patients with MDS were randomly and equally assigned to the treatment group and the control group using a randomized digital table. Thirty patients in each group included 3 risk levels (low, moderate and high risks) with each level 10 patients according to the international prognostic scoring system. The control group was given conventional therapy which was also used in the treatment group. While the treatment group was given Zuogui Pill () and Yougui Pill () for low risk patients; Qingwen Baidu Decoction () and Bazhen Decoction () for moderate risk patients; Gexia Zhuyu Decoction () and Qinghao Biejia Decoction () combined with Shiquan Dabu Decoction () for high risk patients. After the treatment, the differences of overall response rate and immunophenotype (CD13, CD14, CD15, CD33 and CD34) of each group were analyzed. RESULTS The overall response rate of the treatment group was significantly higher than the control group in low risk and moderate risk patients (P=0.029), there was no statistical differences of overall response rate between the treatment group and the control group in high risk patients (P=0.089). The expressions of CD13, CD14, CD33 and CD34 in all three risk levels of the treatment group were obviously decreased after the treatment, while CD15 in all three risk levels of the treatment group was obviously increased after the treatment (P<0.05 or P<0.01). Meanwhile, the difference values of CD13 and CD33 in low risk level of the treatment group, CD33 and CD34 in moderate risk level of the treatment group as well as CD34 and CD15 in high risk level of the treatment group, were all greater than the control groups and they were statistically significant (P<0.05 or P<0.01). CONCLUSIONS It shows a better therapeutic effect if the MDS patients treated with Chinese medicine pattern identification in addition to conventional therapy. Since the treatment may inhibit the malignant clones and improve the dysmaturity of granulocyte differentiation, it is a feasible option in clinical practice.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Hematology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Li-Li Qian
- Department of Hematology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Jian-Ping Shen
- Department of Hematology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Jun-Fa Chen
- Department of Hematology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Yan-Ting Gao
- Department of Hematology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Jing-Jing Xiang
- Department of Hematology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Bao-Dong Ye
- Department of Hematology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Yu-Hong Zhou
- Department of Hematology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, China.
| |
Collapse
|
35
|
Santini V. Treatment of low-risk myelodysplastic syndromes. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2016; 2016:462-469. [PMID: 27913517 PMCID: PMC6142510 DOI: 10.1182/asheducation-2016.1.462] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The majority of myelodysplastic syndrome (MDS) patients belong to the International Prognostic Scoring System (IPSS) and IPSS-revised (IPSS-R) lower-risk categories. Their precise diagnostics and prognostic stratification is often a challenge, but may ensure the optimization of therapy. The availability of diverse treatment options has significantly improved the quality of life and survival of this group of patients. Anemia is the most relevant cytopenia in terms of frequency and symptoms in lower-risk MDS, and may be treated successfully with erythropoietic stimulating agents, provided a careful selection is performed on the basis of IPSS-R, endogenous erythropoietin levels, and transfusion independence. Doses and duration of therapy of erythropoietic-stimulating agents (ESAs) are critical to determine efficacy. In case a patient fails ESA treatment, the available options may include lenalidomide (approved for del5q positive cases), hypomethylating agents, and a rather large number of experimental agents, whose clinical trials should be offered to a larger number of MDS patients. The choice for second-line treatment must take into account biologic, cytogenetic, and molecular-identified characteristics of individual patients, as well as frailty and comorbidities. Other cytopenias are less frequently presenting as isolated. Specific therapy for thrombocytopenia has been proposed in experimental clinical trials with thrombomimetic agents that have shown good efficacy, but raised some safety concern. Although neutropenia is targeted symptomatically with growth factor supportive care, the immunosuppressive treatments are indicated mainly for pancytopenic, hypoplastic lower-risk MDS; they are not widely used because of their toxicity, despite the fact that they may induce responses. Finally, hematopoietic stem cell transplant is the curative option also for lower-risk MDS and timing should be carefully evaluated, balancing toxicity and the possibility of survival advantage. Finally, even when considered suitable for lower-risk MDS, transplant application is limited to the rarer fit and younger MDS patient.
Collapse
Affiliation(s)
- Valeria Santini
- SODc Hematology, Azienda Ospedaliera Universitaria Careggi, University of Florence, Florence, Italy
| |
Collapse
|
36
|
Nairz M, Theurl I, Wolf D, Weiss G. Iron deficiency or anemia of inflammation? : Differential diagnosis and mechanisms of anemia of inflammation. Wien Med Wochenschr 2016; 166:411-423. [PMID: 27557596 PMCID: PMC5065583 DOI: 10.1007/s10354-016-0505-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/30/2016] [Indexed: 02/08/2023]
Abstract
Iron deficiency and immune activation are the two most frequent causes of anemia, both of which are based on disturbances of iron homeostasis. Iron deficiency anemia results from a reduction of the body's iron content due to blood loss, inadequate dietary iron intake, its malabsorption, or increased iron demand. Immune activation drives a diversion of iron fluxes from the erythropoietic bone marrow, where hemoglobinization takes place, to storage sites, particularly the mononuclear phagocytes system in liver and spleen. This results in iron-limited erythropoiesis and anemia. This review summarizes current diagnostic and pathophysiological concepts of iron deficiency anemia and anemia of inflammation, as well as combined conditions, and provides a brief outlook on novel therapeutic options.
Collapse
Affiliation(s)
- Manfred Nairz
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria.
| | - Igor Theurl
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Dominik Wolf
- Medical Clinic III, Department of Oncology, Hematology and Rheumatology, University Clinic Bonn (UKB), Bonn, Germany
| | - Günter Weiss
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria.
| |
Collapse
|
37
|
Santini V, Almeida A, Giagounidis A, Gröpper S, Jonasova A, Vey N, Mufti GJ, Buckstein R, Mittelman M, Platzbecker U, Shpilberg O, Ram R, Del Cañizo C, Gattermann N, Ozawa K, Risueño A, MacBeth KJ, Zhong J, Séguy F, Hoenekopp A, Beach CL, Fenaux P. Randomized Phase III Study of Lenalidomide Versus Placebo in RBC Transfusion-Dependent Patients With Lower-Risk Non-del(5q) Myelodysplastic Syndromes and Ineligible for or Refractory to Erythropoiesis-Stimulating Agents. J Clin Oncol 2016; 34:2988-96. [PMID: 27354480 DOI: 10.1200/jco.2015.66.0118] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE This international phase III, randomized, placebo-controlled, double-blind study assessed the efficacy and safety of lenalidomide in RBC transfusion-dependent patients with International Prognostic Scoring System lower-risk non-del(5q) myelodysplastic syndromes ineligible for or refractory to erythropoiesis-stimulating agents. PATIENTS AND METHODS In total, 239 patients were randomly assigned (2:1) to treatment with lenalidomide (n = 160) or placebo (n = 79) once per day (on 28-day cycles). The primary end point was the rate of RBC transfusion independence (TI) ≥ 8 weeks. Secondary end points were RBC-TI ≥ 24 weeks, duration of RBC-TI, erythroid response, health-related quality of life (HRQoL), and safety. RESULTS RBC-TI ≥ 8 weeks was achieved in 26.9% and 2.5% of patients in the lenalidomide and placebo groups, respectively (P < .001). Ninety percent of patients achieving RBC-TI responded within 16 weeks of treatment. Median duration of RBC-TI with lenalidomide was 30.9 weeks (95% CI, 20.7 to 59.1). Transfusion reduction of ≥ 4 units packed RBCs, on the basis of a 112-day assessment, was 21.8% in the lenalidomide group and 0% in the placebo group. Higher response rates were observed in patients with lower baseline endogenous erythropoietin ≤ 500 mU/mL (34.0% v 15.5% for > 500 mU/mL). At week 12, mean changes in HRQoL scores from baseline did not differ significantly between treatment groups, which suggests that lenalidomide did not adversely affect HRQoL. Achievement of RBC-TI ≥ 8 weeks was associated with significant improvements in HRQoL (P < .01). The most common treatment-emergent adverse events were neutropenia and thrombocytopenia. CONCLUSION Lenalidomide yields sustained RBC-TI in 26.9% of RBC transfusion-dependent patients with lower-risk non-del(5q) myelodysplastic syndromes ineligible for or refractory to erythropoiesis-stimulating agents. Response to lenalidomide was associated with improved HRQoL. Treatment-emergent adverse event data were consistent with the known safety profile of lenalidomide.
Collapse
Affiliation(s)
- Valeria Santini
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ.
| | - Antonio Almeida
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| | - Aristoteles Giagounidis
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| | - Stefanie Gröpper
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| | - Anna Jonasova
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| | - Norbert Vey
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| | - Ghulam J Mufti
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| | - Rena Buckstein
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| | - Moshe Mittelman
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| | - Uwe Platzbecker
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| | - Ofer Shpilberg
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| | - Ron Ram
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| | - Consuelo Del Cañizo
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| | - Norbert Gattermann
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| | - Keiya Ozawa
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| | - Alberto Risueño
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| | - Kyle J MacBeth
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| | - Jianhua Zhong
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| | - Francis Séguy
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| | - Albert Hoenekopp
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| | - C L Beach
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| | - Pierre Fenaux
- Valeria Santini, University of Florence, Florence, Italy; Antonio Almeida, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal; Aristoteles Giagounidis and Stefanie Gröpper, Marien Hospital Düsseldorf; Norbert Gattermann, Heinrich-Heine-Universität, Düsseldorf; Uwe Platzbecker, Technical University Dresden, Dresden, Germany; Anna Jonasova, Charles University General Hospital, Prague, Czech Republic; Norbert Vey, Centre Régional de Lutte Contre le Cancer, Marseille; Pierre Fenaux, Université Paris, Paris, France; Francis Séguy and Albert Hoenekopp, Celgene International, Boudry, Switzerland; Ghulam J. Mufti, King's College Hospital, London, United Kingdom; Rena Buckstein, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Moshe Mittelman and Ron Ram, Tel Aviv University; Ofer Shpilberg, Assuta Medical Center, Tel Aviv, Israel; Consuelo del Cañizo, Hospital Universitario de Salamanca, Salamanca; Alberto Risueño, Celgene Institute for Translational Research Europe, Seville, Spain; Keiya Ozawa, The University of Tokyo, Tokyo, Japan; Kyle J. MacBeth, Celgene Corporation, San Francisco, CA; and Jianhua Zhong and C.L. Beach, Celgene Corporation, Summit, NJ
| |
Collapse
|