1
|
Serrano G, Berastegui N, Díaz-Mazkiaran A, García-Olloqui P, Rodriguez-Res C, Huerga-Dominguez S, Ainciburu M, Vilas-Zornoza A, Martin-Uriz PS, Aguirre-Ruiz P, Ullate-Agote A, Ariceta B, Lamo-Espinosa JM, Acha P, Calvete O, Jimenez T, Molero A, Montoro MJ, Díez-Campelo M, Valcarcel D, Solé F, Alfonso-Pierola A, Ochoa I, Prósper F, Ezponda T, Hernaez M. Single-cell transcriptional profile of CD34+ hematopoietic progenitor cells from del(5q) myelodysplastic syndromes and impact of lenalidomide. Nat Commun 2024; 15:5272. [PMID: 38902243 PMCID: PMC11189937 DOI: 10.1038/s41467-024-49529-x] [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: 11/01/2023] [Accepted: 06/06/2024] [Indexed: 06/22/2024] Open
Abstract
While myelodysplastic syndromes with del(5q) (del(5q) MDS) comprises a well-defined hematological subgroup, the molecular basis underlying its origin remains unknown. Using single cell RNA-seq (scRNA-seq) on CD34+ progenitors from del(5q) MDS patients, we have identified cells harboring the deletion, characterizing the transcriptional impact of this genetic insult on disease pathogenesis and treatment response. Interestingly, both del(5q) and non-del(5q) cells present similar transcriptional lesions, indicating that all cells, and not only those harboring the deletion, may contribute to aberrant hematopoietic differentiation. However, gene regulatory network (GRN) analyses reveal a group of regulons showing aberrant activity that could trigger altered hematopoiesis exclusively in del(5q) cells, pointing to a more prominent role of these cells in disease phenotype. In del(5q) MDS patients achieving hematological response upon lenalidomide treatment, the drug reverts several transcriptional alterations in both del(5q) and non-del(5q) cells, but other lesions remain, which may be responsible for potential future relapses. Moreover, lack of hematological response is associated with the inability of lenalidomide to reverse transcriptional alterations. Collectively, this study reveals transcriptional alterations that could contribute to the pathogenesis and treatment response of del(5q) MDS.
Collapse
Affiliation(s)
- Guillermo Serrano
- Computational Biology Program CIMA-Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), IdISNA, Pamplona, Spain
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Nerea Berastegui
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Aintzane Díaz-Mazkiaran
- Computational Biology Program CIMA-Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), IdISNA, Pamplona, Spain
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Paula García-Olloqui
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Carmen Rodriguez-Res
- Computational Biology Program CIMA-Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), IdISNA, Pamplona, Spain
| | - Sofia Huerga-Dominguez
- Hematology and Cell Therapy Service, Cancer Center Clínica Universidad de Navarra (CCUN), IdISNA, Pamplona, Spain
| | - Marina Ainciburu
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Amaia Vilas-Zornoza
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Patxi San Martin-Uriz
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain
| | - Paula Aguirre-Ruiz
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain
| | - Asier Ullate-Agote
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain
| | - Beñat Ariceta
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | | | - Pamela Acha
- MDS Research Group, Josep Carreras Leukaemia Research Institut, Universitat Autònoma de Barcelona, Barcelona, Spain
- Service of Hematology, Hospital Universitari Vall d'Hebron, Barcelona; Vall d'Hebron Instituto de Oncología (VHIO), Barcelona, Spain
| | - Oriol Calvete
- MDS Research Group, Josep Carreras Leukaemia Research Institut, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Tamara Jimenez
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
- Department of Hematology, Hospital Universitario de Salamanca-IBSAL, Salamanca, Spain
| | - Antonieta Molero
- Service of Hematology, Hospital Universitari Vall d'Hebron, Barcelona; Vall d'Hebron Instituto de Oncología (VHIO), Barcelona, Spain
| | - Maria Julia Montoro
- Service of Hematology, Hospital Universitari Vall d'Hebron, Barcelona; Vall d'Hebron Instituto de Oncología (VHIO), Barcelona, Spain
| | - Maria Díez-Campelo
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
- Department of Hematology, Hospital Universitario de Salamanca-IBSAL, Salamanca, Spain
| | - David Valcarcel
- Service of Hematology, Hospital Universitari Vall d'Hebron, Barcelona; Vall d'Hebron Instituto de Oncología (VHIO), Barcelona, Spain
| | - Francisco Solé
- MDS Research Group, Josep Carreras Leukaemia Research Institut, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ana Alfonso-Pierola
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
- Hematology and Cell Therapy Service, Cancer Center Clínica Universidad de Navarra (CCUN), IdISNA, Pamplona, Spain
| | - Idoia Ochoa
- Instituto de Ciencia de los Datos e Inteligencia Artificial (DATAI), University of Navarra, Pamplona, Spain
- Department of Electrical and Electronics engineering, School of Engineering (Tecnun), University of Navarra, Donostia, Spain
| | - Felipe Prósper
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain.
- Hematology and Cell Therapy Service, Cancer Center Clínica Universidad de Navarra (CCUN), IdISNA, Pamplona, Spain.
| | - Teresa Ezponda
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain.
| | - Mikel Hernaez
- Computational Biology Program CIMA-Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), IdISNA, Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain.
- Instituto de Ciencia de los Datos e Inteligencia Artificial (DATAI), University of Navarra, Pamplona, Spain.
| |
Collapse
|
2
|
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
|
3
|
Abdulbaki R, Pullarkat ST. A Brief Overview of the Molecular Landscape of Myelodysplastic Neoplasms. Curr Oncol 2024; 31:2353-2363. [PMID: 38785456 PMCID: PMC11119831 DOI: 10.3390/curroncol31050175] [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: 03/13/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
Myelodysplastic neoplasm (MDS) is a heterogeneous group of clonal hematological disorders that originate from the hematopoietic and progenitor cells and present with cytopenias and morphologic dysplasia with a propensity to progress to bone marrow failure or acute myeloid leukemia (AML). Genetic evolution plays a critical role in the pathogenesis, progression, and clinical outcomes of MDS. This process involves the acquisition of genetic mutations in stem cells that confer a selective growth advantage, leading to clonal expansion and the eventual development of MDS. With the advent of next-generation sequencing (NGS) assays, an increasing number of molecular aberrations have been discovered in recent years. The knowledge of molecular events in MDS has led to an improved understanding of the disease process, including the evolution of the disease and prognosis, and has paved the way for targeted therapy. The 2022 World Health Organization (WHO) Classification and the International Consensus Classification (ICC) have incorporated the molecular signature into the classification system for MDS. In addition, specific germline mutations are associated with MDS development, especially in pediatrics and young adults. This article reviews the genetic abnormalities of MDS in adults with a brief review of germline predisposition syndromes.
Collapse
Affiliation(s)
- Rami Abdulbaki
- Department of Pathology, Laboratory Medicine, UCLA, David Geffen School of Medicine, Los Angeles, CA 90095, USA;
| | | |
Collapse
|
4
|
Lawal RA, Banjoko O, Ndulue C, Adebeshin ST, Sharif A, Ighodaro OE, Olusoji R, Odusanya B, El-Hamdi NS. Improved Side Effect Profile of Alternate-Day Dosing of Lenalidomide. Cureus 2024; 16:e55317. [PMID: 38559519 PMCID: PMC10981840 DOI: 10.7759/cureus.55317] [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] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
Myelodysplastic syndrome (MDS) is a heterogeneous hematological condition associated with cytopenia, inadequate blood cell synthesis, and the risk of developing acute myeloid leukemia (AML). Patients are divided into risk groups according to the International Prognostic Scoring System (IPSS) to help direct therapy. Allogeneic stem cell transplantation, despite its limitations, is curative. Medical management, such as the use of lenalidomide, has potential benefits but can cause adverse effects that require dose regimen modification. Lenalidomide is approved for low-risk MDS with 5q deletion (5q- MDS). In this case study, a 79-year-old woman with 5q- MDS was switched from a daily regimen to an alternate-day lenalidomide dose schedule to achieve complete remission with fewer adverse effects. The management of hematological toxicity and the mechanisms of action of lenalidomide are discussed. We recommend individualized treatment strategies and additional research to improve MDS management.
Collapse
Affiliation(s)
- Ridwan A Lawal
- Internal Medicine, College of Medicine, University of Lagos, Lagos, NGA
| | - Oluwole Banjoko
- Internal Medicine, Lagos State University Teaching Hospital, Lagos, NGA
| | | | | | - Arsalan Sharif
- Medicine, Tbilisi State Medical University, Tbilisi, GEO
| | | | - Rahman Olusoji
- Internal Medicine, Columbia University at Harlem Hospital Center, New York, USA
| | | | - Nadia S El-Hamdi
- Internal Medicine, Hospital Corporation of America (HCA) Houston Healthcare Kingwood, University of Houston College of Medicine (UHCOM), Texas, USA
| |
Collapse
|
5
|
Elkholy MM, Fahmi MW, El-Haggar SM. Dynamic changes in the levels of sCD62L and SPARC in chronic myeloid leukaemia patients during imatinib treatment. J Clin Pharm Ther 2022; 47:2115-2129. [PMID: 36053969 DOI: 10.1111/jcpt.13759] [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: 06/11/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 12/29/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Chronic myeloid leukaemia (CML) microenvironment is responsible for resistance of leukaemic cells to tyrosine kinase inhibitor, altered adhesion, increased proliferation and leukaemic cells growth and survival through the secretion of many soluble molecules. We aimed at monitoring soluble L-selectin (sCD62L) and secreted protein acidic and rich in cysteine (SPARC) levels in chronic phase chronic myeloid leukaemia (CP-CML) patients and assessing the impact of imatinib on these parameters. METHODS This prospective controlled clinical trial enrolled 35 subjects classified into two groups: control group included 10 healthy volunteers and CP-CML patients group included 25 newly diagnosed CP-CML patients received imatinib 400 mg once daily. sCD62L plasma levels, SPARC serum levels, breakpoint cluster region-Abelson1 (BCR-ABL1) %, complete blood count with differential, liver and kidney functions parameters were assessed at baseline and after 3 and 6 months of treatment. RESULTS AND DISCUSSION At baseline, sCD62L and SPARC were significantly elevated in CP-CML patients (p < 0.05) compared to control group. After 3 months of treatment, sCD62L was non-significantly decreased (p > 0.05), while surprisingly SPARC was significantly increased (p < 0.05) compared to baseline. Moreover, after 6 months of treatment, sCD62L was significantly decreased (p < 0.05) and SPARC was non-significantly decreased (p > 0.05) compared to baseline. In addition, sCD62L was significantly correlated with WBCs and neutrophils counts, while SPARC was significantly correlated with lymphocytes count at baseline and after 3 and 6 months of imatinib treatment. WHAT IS NEW AND CONCLUSION The elevated levels of sCD62L and SPARC at diagnosis in CP-CML patients could reflect their roles in CML pathogenesis and the dynamic changes in their levels during imatinib therapy might suppose additional mechanisms of action of imatinib beside inhibition of BCR-ABL. Furthermore, imatinib showed a significant impact on sCD62L and SPARC levels during treatment period.
Collapse
Affiliation(s)
- Mahmoud Mohamed Elkholy
- Clinical Pharmacy Department, Faculty of Pharmacy, Al Salam University in Egypt, Kafr El-Zayat, Egypt
| | - Maryan Waheeb Fahmi
- Medical Oncology Unit, Internal Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | | |
Collapse
|
6
|
Nian Q, Li J, Han Z, Liang Q, Liu M, Yang C, Rodrigues-Lima F, Jiang T, Zhao L, Zeng J, Liu C, Shi J. SPARC in hematologic malignancies and novel technique for hematological disease with its abnormal expression. Biomed Pharmacother 2022; 153:113519. [DOI: 10.1016/j.biopha.2022.113519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 11/27/2022] Open
|
7
|
Aly NAR, Rizk S, Aboul Enein A, El Desoukey N, Zawam H, Ahmed M, El Shikh ME, Pitzalis C. The role of lymphoid tissue SPARC in the pathogenesis and response to treatment of multiple myeloma. Front Oncol 2022; 12:1009993. [PMID: 36605435 PMCID: PMC9807864 DOI: 10.3389/fonc.2022.1009993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/25/2022] [Indexed: 12/24/2022] Open
Abstract
Background Despite the significant progress in the treatment of multiple myeloma (MM), the disease remains untreatable and its cure is still an unmet clinical need. Neoplastic transformation in MM is initiated in the germinal centers (GCs) of secondary lymphoid tissue (SLT) where B cells experience extensive somatic hypermutation induced by follicular dendritic cells (FDCs) and T-cell signals. Objective We reason that secreted protein acidic and rich in cysteine (SPARC), a common stromal motif expressed by FDCs at the origin (SLTs) and the destination (BM) of MM, plays a role in the pathogenesis of MM, and, here, we sought to investigate this role. Methods There were 107 BM biopsies from 57 MM patients (taken at different time points) together with 13 control specimens assessed for SPARC gene and protein expression and compared with tonsillar tissues. In addition, regulation of myeloma-promoting genes by SPARC-secreting FDCs was assessed in in vitro GC reactions (GCRs). Results SPARC gene expression was confirmed in both human primary (BM) and secondary (tonsils) lymphoid tissues, and the expression was significantly higher in the BM. Sparc was detectable in the BM and tonsillar lysates, co-localized with the FDC markers in both tissues, and stimulation of FDCs in vitro induced significantly higher levels of SPARC expression than unstimulated controls. In addition, SPARC inversely correlated with BM PC infiltration, ISS staging, and ECOG performance of the MM patients, and in vitro addition of FDCs to lymphocytes inhibited the expression of several oncogenes associated with malignant transformation of PCs. Conclusion FDC-SPARC inhibits several myelomagenic gene expression and inversely correlates with PC infiltration and MM progression. Therapeutic induction of SPARC expression through combinations of the current MM drugs, repositioning of non-MM drugs, or novel drug discovery could pave the way to better control MM in clinically severe and drug-resistant patients.
Collapse
Affiliation(s)
- Nesreen Amer Ramadan Aly
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Samia Rizk
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Azza Aboul Enein
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nermeen El Desoukey
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Hamdy Zawam
- Clinical Oncology and Nuclear Radiation Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Manzoor Ahmed
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Mohey Eldin El Shikh
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- *Correspondence: Mohey Eldin El Shikh,
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
8
|
Cencini E, Fabbri A, Mecacci B, Bocchia M. Role of lenalidomide in the treatment of peripheral T-cell non-Hodgkin lymphomas. World J Clin Oncol 2021; 12:882-896. [PMID: 34733611 PMCID: PMC8546656 DOI: 10.5306/wjco.v12.i10.882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/07/2021] [Accepted: 09/02/2021] [Indexed: 02/06/2023] Open
Abstract
T-cell lymphomas (TCLs) represent a group of lymphoid neoplasms characterized by an aggressive clinical course, even after an anthracycline-containing regimen. Novel agents for patients with relapsed/refractory TCL are urgently needed. Lenalidomide is an oral drug with immunomodulatory, antiangiogenic and direct antineoplastic effects. These peculiar mechanisms of action make TCL an attractive target for lenalidomide. We have identified five clinical trials in which lenalidomide monotherapy was investigated to treat TCL, including cutaneous TCL (CTCL) and adult T-cell lymphoma/leukemia (ATLL). In the ATLL-002 study, the overall response rate (ORR) was 42% and median progression-free survival (PFS) and overall survival were 3.8 mo and 20.3 mo, respectively. In a phase II trial for CTCL, ORR was 28% and median PFS and overall survival were 8 mo and 43 mo, respectively. For nodal peripheral TCL, ORR was between 10% and 43% in three clinical trials, with a median PFS of about 4 mo, even if some patients had a durable response. Overall toxicity is manageable and grade 3-4 events are mainly hematological and reversible. Combination strategies did not improve PFS. In conclusion, lenalidomide could represent a suitable treatment option for relapsed/refractory TCL, especially for neoplasms with a T-follicular helper origin, such as angioimmunoblastic TCL.
Collapse
Affiliation(s)
- Emanuele Cencini
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese and University of Siena, Siena 53100, Italy
| | - Alberto Fabbri
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese and University of Siena, Siena 53100, Italy
| | - Bianca Mecacci
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese and University of Siena, Siena 53100, Italy
| | - Monica Bocchia
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese and University of Siena, Siena 53100, Italy
| |
Collapse
|
9
|
Park I, Phan TM, Fang J. Novel Molecular Mechanism of Lenalidomide in Myeloid Malignancies Independent of Deletion of Chromosome 5q. Cancers (Basel) 2021; 13:5084. [PMID: 34680233 PMCID: PMC8534127 DOI: 10.3390/cancers13205084] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 09/30/2021] [Accepted: 10/08/2021] [Indexed: 12/26/2022] Open
Abstract
Lenalidomide as well as other immunomodulatory drugs (IMiDs) have achieved clinical efficacies in certain sub-types of hematologic malignancies, such as multiple myeloma, lower-risk myelodysplastic syndromes (MDS) with a single deletion of chromosome 5q (del(5q)) and others. Despite superior clinical response to lenalidomide in hematologic malignancies, relapse and resistance remains a problem in IMiD-based therapy. The last ten years have witnessed the discovery of novel molecular mechanism of IMiD-based anti-tumor therapy. IMiDs bind human cereblon (CRBN), the substrate receptor of the CRL4 E3 ubiquitin ligase complex. Binding of CRBN with IMiDs leads to degradation of the Ikaros family zinc finger proteins 1 and 3 (IKZF1 and IKZF3) and casein kinase 1 alpha. We have found that lenalidomide-mediated degradation of IKZF1 leads to activation of the G protein-coupled receptor 68 (GPR68)/calcium/calpain pro-apoptotic pathway and inhibition of the regulator of calcineurin 1 (RCAN1)/calcineurin pro-survival pathway in MDS and acute myeloid leukemia (AML). Calcineurin inhibitor Cyclosporin-A potentiates the anti-leukemia activity of lenalidomide in MDS/AML with or without del(5q). These findings broaden the therapeutic potential of IMiDs. This review summarizes novel molecular mechanism of lenalidomide in myeloid malignancies, especially without del(5q), in the hope to highlight novel therapeutic targets.
Collapse
Affiliation(s)
| | | | - Jing Fang
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC 29208, USA; (I.P.); (T.M.P.)
| |
Collapse
|
10
|
Sundaresan L, Giri S, Singh H, Chatterjee S. Repurposing of thalidomide and its derivatives for the treatment of SARS-coV-2 infections: Hints on molecular action. Br J Clin Pharmacol 2021; 87:3835-3850. [PMID: 33609410 PMCID: PMC8013920 DOI: 10.1111/bcp.14792] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 01/27/2021] [Accepted: 02/08/2021] [Indexed: 01/08/2023] Open
Abstract
AIMS The SARS-coV-2 pandemic continues to cause an unprecedented global destabilization requiring urgent attention towards drug and vaccine development. Thalidomide, a drug with known anti-inflammatory and immunomodulatory effects has been indicated to be effective in treating a SARS-coV-2 pneumonia patient. Here, we study the possible mechanisms through which thalidomide might affect coronavirus disease-19 (COVID-19). METHODS The present study explores the possibility of repurposing thalidomide for the treatment of SARS-coV-2 pneumonia by reanalysing transcriptomes of SARS-coV-2 infected tissues with thalidomide and lenalidomide induced transcriptomic changes in transformed lung and haematopoietic models as procured from databases, and further comparing them with the transcriptome of primary endothelial cells. RESULTS Thalidomide and lenalidomide exhibited pleiotropic effects affecting a range of biological processes including inflammation, immune response, angiogenesis, MAPK signalling, NOD-like receptor signalling, Toll-like receptor signalling, leucocyte differentiation and innate immunity, the processes that are aberrantly regulated in severe COVID-19 patients. CONCLUSION The present study indicates thalidomide analogues as a better fit for treating severe cases of novel viral infections, healing the damaged network by compensating the impairment caused by the COVID-19.
Collapse
Affiliation(s)
| | - Suvendu Giri
- Vascular Biology LaboratoryAU‐KBC Research CentreChennaiIndia
- Department of BiotechnologyAnna UniversityChennaiIndia
| | - Himanshi Singh
- Vascular Biology LaboratoryAU‐KBC Research CentreChennaiIndia
- Department of BiotechnologyAnna UniversityChennaiIndia
| | - Suvro Chatterjee
- Vascular Biology LaboratoryAU‐KBC Research CentreChennaiIndia
- Department of BiotechnologyAnna UniversityChennaiIndia
| |
Collapse
|
11
|
Chen Y, Talukder R, Merritt BY, King KY, Kimmel M, Rivero G, Sosa R. Genomic trajectory in leukemogenesis of myeloproliferative neoplasms: a case report. BMC Med Genomics 2021; 14:137. [PMID: 34022887 PMCID: PMC8141236 DOI: 10.1186/s12920-021-00986-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 05/18/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND We report a patient with Essential Thrombocythemia (ET), subsequently diagnosed with concurrent myeloid and lymphoid leukemia. Generally, the molecular mechanisms underlying leukemic transformation of Philadelphia-negative myeloproliferative neoplasms (Ph-MPN) are poorly understood. Risk of transformation to acute myelogenous leukemia (AML) is low; transformation to both AML and acute lymphoblastic leukemia (ALL) is extremely low. Genetic defects, including allele burden, order of mutation acquisition, clonal heterogeneity and epigenetic mechanisms are important contributors to disease acceleration. CASE PRESENTATION A 78-year-old Caucasian female originally treated for stable ET, underwent disease acceleration and transition to myeloid sarcoma and B-cell ALL. Genomic reconstruction based on targeted sequencing revealed the presence of a large del(5q) in all three malignancies and somatic driver mutations: TET2, TP53, SF3B1, and ASXL1 at high allele frequency. We propose that the combination of genetic and molecular abnormalities led to hematopoietic stem cell (HSC) injury and disease progression through sub-clone branching. We hypothesize that ancestral reconstruction of genomic data is a useful tool to uncover subclonal events leading to transformation. CONCLUSIONS The use of ancestral reconstruction of genomic data sheds light on the unique clinical scenario described in this case report. By determining the mutational profile of tumors at several timepoints and deducing the most parsimonious relationship between them, we propose a reconstruction of their origin. We propose that blast progression originated from subclonal events with malignant potential, which coexisted with but did not originate from JAK2 p.V617F-positive ET. We conclude that the application of genomic reconstruction enhances our understanding of leukemogenesis by identifying the timing of molecular events, potentially leading to better chemotherapy choices as well as the development of new targeted therapies.
Collapse
Affiliation(s)
- Yujie Chen
- Department of Statistics and Bioengineering, Rice University, 6100 Main Street, Houston, TX, USA
| | - Rafee Talukder
- Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Brian Y Merritt
- The Dan L. Duncan Comprehensive Cancer Center at Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Department of Pathology and Immunology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor Genetics and Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Katherine Y King
- The Dan L. Duncan Comprehensive Cancer Center at Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Department of Pediatrics, Section of Infectious Disease, Baylor College of Medicine, 1102 Bates St. Suite 1150, Houston, TX, USA
| | - Marek Kimmel
- Department of Statistics and Bioengineering, Rice University, 6100 Main Street, Houston, TX, USA
| | - Gustavo Rivero
- Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA, 19111, USA
- Section of Hematology and Oncology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Romina Sosa
- The Dan L. Duncan Comprehensive Cancer Center at Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA.
- Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA, 19111, USA.
- Section of Hematology and Oncology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA.
| |
Collapse
|
12
|
Zhou Y, Tan Z, Xue P, Wang Y, Li X, Guan F. High-throughput, in-depth and estimated absolute quantification of plasma proteome using data-independent acquisition/mass spectrometry ("HIAP-DIA"). Proteomics 2021; 21:e2000264. [PMID: 33460299 DOI: 10.1002/pmic.202000264] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/07/2021] [Accepted: 01/07/2021] [Indexed: 01/01/2023]
Abstract
Mass spectrometry-based plasma proteomics has been demonstrated to be a useful tool capable of quantifying hundreds of proteins in a single LC-MS/MS experiment, for biomarker discovery or elucidation of disease mechanisms. We developed a novel data-independent acquisition (DIA)/MS-based workflow for high-throughput, in-depth and estimated absolute quantification of plasma proteins (termed HIAP-DIA), without depleting high-abundant proteins, in a single-shot experiment. In HIAP-DIA workflow, we generated an ultra-deep cumulative undepleted and depleted spectral library which contained 55,157 peptides and 5,328 proteins, optimized column length (50 cm) and gradient (90 min) of liquid chromatography instrumentation, optimized 50 DIA segments with average isolation window 17 Th, and selected reference proteins for estimated absolute quantification of all plasma proteins. A total of 606 proteins were quantified in triplicate, and 427 proteins were quantified with CV <20% in plasma proteome. R-squared value of overlapped 208 endogenous PQ500 estimated protein amounts from HIAP-DIA and absolute quantification with internal standards was 0.82, indicating high quantification accuracy of HIAP-DIA. As a pilot study, the HIAP-DIA approach described here was applied to a myelodysplastic syndromes (MDS) disease cohort. We achieved absolute quantification of 789 plasma proteins in 22 clinical plasma samples, spanning less than six orders of magnitude with quantification limit 10-20 ng/mL, and discovered 95 differentially expressed proteins providing insights into MDS pathophysiology.
Collapse
Affiliation(s)
- Yue Zhou
- The Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Zengqi Tan
- College of Life Science, Northwest University, Xi'an, China
| | - Peng Xue
- Department of Biology, Institute of Molecular Systems Biology, Zürich, Switzerland
| | - Yi Wang
- Department of Hematology, Provincial People's Hospital, Xi'an, China
| | - Xiang Li
- College of Life Science, Northwest University, Xi'an, China
| | - Feng Guan
- The Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China.,College of Life Science, Northwest University, Xi'an, China
| |
Collapse
|
13
|
Janovská P, Normant E, Miskin H, Bryja V. Targeting Casein Kinase 1 (CK1) in Hematological Cancers. Int J Mol Sci 2020; 21:E9026. [PMID: 33261128 PMCID: PMC7730698 DOI: 10.3390/ijms21239026] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/20/2020] [Accepted: 11/21/2020] [Indexed: 02/06/2023] Open
Abstract
The casein kinase 1 enzymes (CK1) form a family of serine/threonine kinases with seven CK1 isoforms identified in humans. The most important substrates of CK1 kinases are proteins that act in the regulatory nodes essential for tumorigenesis of hematological malignancies. Among those, the most important are the functions of CK1s in the regulation of Wnt pathways, cell proliferation, apoptosis and autophagy. In this review we summarize the recent developments in the understanding of biology and therapeutic potential of the inhibition of CK1 isoforms in the pathogenesis of chronic lymphocytic leukemia (CLL), other non-Hodgkin lymphomas (NHL), myelodysplastic syndrome (MDS), acute myeloid leukemia (AML) and multiple myeloma (MM). CK1δ/ε inhibitors block CLL development in preclinical models via inhibition of WNT-5A/ROR1-driven non-canonical Wnt pathway. While no selective CK1 inhibitors have reached clinical stage to date, one dual PI3Kδ and CK1ε inhibitor, umbralisib, is currently in clinical trials for CLL and NHL patients. In MDS, AML and MM, inhibition of CK1α, acting via activation of p53 pathway, showed promising preclinical activities and the first CK1α inhibitor has now entered the clinical trials.
Collapse
Affiliation(s)
- Pavlína Janovská
- Department of Experimental Biology, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic;
| | | | - Hari Miskin
- TG Therapeutics, New York, NY 10014, USA; (E.N.); (H.M.)
| | - Vítězslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic;
- Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of the Czech Republic, 61265 Brno, Czech Republic
| |
Collapse
|
14
|
Dou A, Fang J. Cyclosporine Broadens the Therapeutic Potential of Lenalidomide in Myeloid Malignancies. JOURNAL OF CELLULAR IMMUNOLOGY 2020; 2:237-244. [PMID: 32984863 PMCID: PMC7518522 DOI: 10.33696/immunology.2.049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The immunomodulatory drug lenalidomide is used for the treatment of certain hematologic malignancies, including myelodysplastic syndromes (MDS). Lenalidomide interacts with cereblon (CRBN), a component of the CRL4CRBN E3 ubiquitin ligase complex, leading to ubiquitination and subsequent degradation of substrates, such as transcription factor Ikaros (Ikaros family zinc finger 1, IKZF1). With a genome loss of function screen, we recently identified two novel pathways mediated by lenalidomide in MDS. In this review, we summarized the major findings of these two pathways and their clinical implications. Depletion of G protein-coupled receptor 68 (GPR68) or an endogenous calcineurin (CaN) inhibitor, regulator of calcineurin 1 (RCAN1), reversed the inhibitory effect of lenalidomide on MDSL cells, an MDS cell line. Intriguingly, both GPR68 and RCAN1 expression levels were upregulated in MDSL cells after treatment with lenalidomide that was dependent on diminishment of IKZF1, indicating that IKZF1 functioned as a transcription repressor for GPR68 and RCAN1. Mechanistic studies revealed that upregulation or activation of GPR68 induced a Ca2+/calpain pro-apoptotic pathway, while upregulation of RCAN1 inhibited the CaN pro-survival pathway in MDSL cells. Notably, the pharmacological CaN inhibitor, cyclosporine, enhanced the sensitivity to lenalidomide in MDS as well as acute myeloid leukemia (AML). Surprisingly, pretreatment with lenalidomide reversed the immunosuppressive effects of cyclosporine on T lymphocytes. Our studies suggest that lenalidomide mediates degradation of IKZF1, leading to derepression of GPR68 and RCAN1 that activates the Ca2+/calpain pro- apoptotic pathway and inhibits the CaN pro-survival pathway, respectively. Our studies implicate that cyclosporine extends the therapeutic potential of lenalidomide to myeloid malignancies without compromising immune function.
Collapse
Affiliation(s)
- Aixia Dou
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC, USA
| | - Jing Fang
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC, USA
| |
Collapse
|
15
|
Ma LY, Su L. Application of Lenalidomide on Diffused Large B-cell Lymphoma: Salvage, Maintenance, and Induction Treatment. Chin Med J (Engl) 2019; 131:2510-2513. [PMID: 30334543 PMCID: PMC6202603 DOI: 10.4103/0366-6999.243567] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Li-Yangxue Ma
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Li Su
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|
16
|
Mährle T, Akyüz N, Fuchs P, Bonzanni N, Simnica D, Germing U, Asemissen AM, Jann JC, Nolte F, Hofmann WK, Nowak D, Binder M. Deep sequencing of bone marrow microenvironments of patients with del(5q) myelodysplastic syndrome reveals imprints of antigenic selection as well as generation of novel T-cell clusters as a response pattern to lenalidomide. Haematologica 2019; 104:1355-1364. [PMID: 30655375 PMCID: PMC6601099 DOI: 10.3324/haematol.2018.208223] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 01/15/2019] [Indexed: 12/11/2022] Open
Abstract
In myelodysplastic syndromes with a partial deletion of the long arm of chromosome 5, del(5q), lenalidomide is believed to reverse anergic T-cell immunity in the bone marrow resulting in suppression of the del(5q) clone. In this study we used next-generation sequencing of immunoglobulin heavy chain (IGH) and T-cell receptor beta (TRB) rearrangements in bone marrow-residing and peripheral blood-circulating lymphocytes of patients with del(5q) myelodysplastic syndromes to assess the immune architecture and track adaptive immune responses during treatment with lenalidomide. The baseline bone marrow B-cell space in patients was comparable to that of age-matched healthy controls in terms of gene usage and IGH clonality, but showed a higher percentage of hypermutated IGH sequences, indicating an expanded number of antigen-experienced B lineage cells. Bone marrow B lineage clonality decreased significantly and hypermutated IGH clones normalized upon lenalidomide treatment, well in line with the proliferative effect on healthy antigen-inexperienced B-cell precursors previously described for this drug. The T-cell space in bone marrow of patients with del(5q) myelodysplastic syndromes showed higher TRB clonality compared to that of healthy controls. Upon lenalidomide treatment, myelodysplastic syndrome-specific T-cell clusters with low to medium spontaneous generation probabilities emerged; these clusters were shared across patients, indicating a common antigen-driven T-cell response pattern. Hence, we observed B lineage diversification and generation of new, antigen-dependent T-cell clusters, compatible with a model of adaptive immunity induced against the del(5q) clone by lenalidomide. Overall, this supports the concept that lenalidomide not only alters the functional T-cell state, but also the composition of the T- and B-cell repertoires in del(5q) myelodysplastic syndromes.
Collapse
Affiliation(s)
- Thorben Mährle
- Department of Oncology and Hematology, BMT with Pneumology section, Hubertus Wald Tumorzentrum / UCCH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nuray Akyüz
- Department of Oncology and Hematology, BMT with Pneumology section, Hubertus Wald Tumorzentrum / UCCH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Pim Fuchs
- ENPICOM, 's-Hertogenbosch, the Netherlands
| | | | - Donjete Simnica
- Department of Oncology and Hematology, BMT with Pneumology section, Hubertus Wald Tumorzentrum / UCCH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Hematology and Oncology, University Hospital Halle (Saale), Germany
| | - Ulrich Germing
- Department of Hematology, Oncology, and Clinical Immunology, Heinrich Heine University Düsseldorf, Germany
| | - Anne Marie Asemissen
- Department of Oncology and Hematology, BMT with Pneumology section, Hubertus Wald Tumorzentrum / UCCH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johann Christoph Jann
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Florian Nolte
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Daniel Nowak
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Mascha Binder
- Department of Oncology and Hematology, BMT with Pneumology section, Hubertus Wald Tumorzentrum / UCCH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany .,Department of Hematology and Oncology, University Hospital Halle (Saale), Germany
| |
Collapse
|
17
|
Lee JH, List A, Sallman DA. Molecular pathogenesis of myelodysplastic syndromes with deletion 5q. Eur J Haematol 2019; 102:203-209. [PMID: 30578738 DOI: 10.1111/ejh.13207] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 12/08/2018] [Accepted: 12/10/2018] [Indexed: 12/16/2022]
Abstract
The molecular pathogenesis of deletion 5q (del(5q)) myelodysplastic syndrome (MDS) has recently been realized as a result of major advances in our understanding of the mechanisms responsible for clinical phenotype. Identification of commonly deleted genes such as RPS14, miRNA-145, HSPA9, CD78, and CSNK1a1 have elucidated the precise biological changes responsible for the anemia, leukopenia, and thrombocytosis that characterizes del(5q) MDS and highlighted the importance of allelic haploinsufficiency in the hematological phenotype. Recent elegant investigations have also identified a critical role of innate immune signaling in del(5q) pathogenesis. TP53 and Wnt/β-catenin pathways have also been found to be involved in clonal expansion and progression of the disease as well as resistance and poor outcomes to available therapy. Understanding the molecular pathogenesis of the disease has provided a critical foundation in identifying the biological targets of lenalidomide in del(5q) MDS, which has led to the development of novel therapeutic agents in hematologic malignancies as well as potential alternative targets to exploit in patients who have failed lenalidomide treatment.
Collapse
Affiliation(s)
- Jung-Hoon Lee
- University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Alan List
- Malignant Hematology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - David A Sallman
- Malignant Hematology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
18
|
SOHO State of the Art and Next Questions: Management of Myelodysplastic Syndromes With Deletion 5q. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2018; 18:629-635. [DOI: 10.1016/j.clml.2018.07.293] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 07/25/2018] [Indexed: 12/17/2022]
|
19
|
Hosono N, Makishima H, Mahfouz R, Przychodzen B, Yoshida K, Jerez A, LaFramboise T, Polprasert C, Clemente MJ, Shiraishi Y, Chiba K, Tanaka H, Miyano S, Sanada M, Cui E, Verma AK, McDevitt MA, List AF, Saunthararajah Y, Sekeres MA, Boultwood J, Ogawa S, Maciejewski JP. Recurrent genetic defects on chromosome 5q in myeloid neoplasms. Oncotarget 2018; 8:6483-6495. [PMID: 28031539 PMCID: PMC5351647 DOI: 10.18632/oncotarget.14130] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 12/16/2016] [Indexed: 11/25/2022] Open
Abstract
Background Deletion of chromosome 5q (del(5q)) is the most common karyotypic abnormality in myeloid neoplasms. Materials and Methods To define the pathogenic molecular features associated with del(5q), next–generation sequencing was applied to 133 patients with myeloid neoplasms (MDS; N = 69, MDS/MPN; N = 5, sAML; N = 29, pAML; N = 30) with del(5q) as a sole abnormally or a part of complex karyotype and results were compared to molecular features of patients diploid for chr5. Findings A number of 5q genes with haploinsufficient expression and/or recurrent somatic mutations were identified; for these genes, CSNK1A1 and G3BP1 within the commonly deleted 5q region and DDX41 within a commonly retained region were most commonly affected by somatic mutations. These genes showed consistent haploinsufficiency in deleted cases; low expression/mutations of G3BP1 or DDX41 were associated with poor survival, likely due to decreased cellular function. The most common mutations on other chromosomes in patients with del(5q) included TP53, and mutations of FLT3 (ITD or TKD), NPM1 or TET2 and were mutually exclusive. Serial sequencing allowed for definition of clonal architecture and dynamics, in patients with exome sequencing allelic imbalance for informative SNPs facilitated simultaneous approximation of clonal size of del(5q) and clonal burden for somatic mutations. Interpretation Our results illuminate the spectrum of molecular defects characteristic of del(5q), their clinical impact and succession of stepwise evolution.
Collapse
Affiliation(s)
- Naoko Hosono
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA.,First Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Hideki Makishima
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Reda Mahfouz
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Bartlomiej Przychodzen
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kenichi Yoshida
- Cancer Genomics Project, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Andres Jerez
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Thomas LaFramboise
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Chantana Polprasert
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Michael J Clemente
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Yuichi Shiraishi
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kenichi Chiba
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hiroko Tanaka
- Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Satoru Miyano
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masashi Sanada
- Cancer Genomics Project, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Edward Cui
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Amit K Verma
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Michael A McDevitt
- Division of Hematology and Hematological Malignancy, Department of Internal Medicine and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alan F List
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Yogen Saunthararajah
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Mikkael A Sekeres
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA.,Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA.,LLR Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Jacqueline Boultwood
- LLR Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Seishi Ogawa
- Cancer Genomics Project, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Jaroslaw P Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA.,Leukemia Program, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH, USA
| |
Collapse
|
20
|
Guo J, Fei C, Zhao Y, Zhao S, Zheng Q, Su J, Wu D, Li X, Chang C. Lenalidomide restores the osteogenic differentiation of bone marrow mesenchymal stem cells from multiple myeloma patients via deactivating Notch signaling pathway. Oncotarget 2017; 8:55405-55421. [PMID: 28903429 PMCID: PMC5589668 DOI: 10.18632/oncotarget.19265] [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: 02/14/2017] [Accepted: 06/24/2017] [Indexed: 01/01/2023] Open
Abstract
Multiple myeloma (MM) always presents osteolytic bone lesions, resulting from the abnormal osteoblastic and osteoclastic function in patients. MM patients exhibit the impairment of osteogenic differentiation of BMMSCs (bone marrow mesenchymal stem cells) and osteoblast deficiency. Effects of the drug, lenalidomide on the osteoblastic functions and the involved mechanisms remain unexplored. In the present study, it is observed that the osteogenic differentiation of BMMSCs from MM patients (MM-MSCs) is impaired and activation of Notch signaling pathway in MM-MSCs is abnormal. Notch signaling activation inhibits BMMSCs osteogenesis. Knockdown of Notch1 expression and DAPT application reverse the osteogenic differentiation from MM-MSCs. Furthermore, it is shown that the gene expression of Notch signaling molecules, including receptors, ligands and downstream factors are significantly decreased in MM-MSCs following lenalidomide treatment, compared with non-treated MM-MSCs. Taken together, treatment with lenalidomide restores the osteogenic differentiation of MM-MSCs via deactivating Notch signaling pathway.
Collapse
Affiliation(s)
- Juan Guo
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Chengming Fei
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Youshan Zhao
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Sida Zhao
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Qingqing Zheng
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Jiying Su
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Dong Wu
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Xiao Li
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Chunkang Chang
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
21
|
Talati C, Sallman D, List A. Lenalidomide: Myelodysplastic syndromes with del(5q) and beyond. Semin Hematol 2017; 54:159-166. [PMID: 28958290 DOI: 10.1053/j.seminhematol.2017.06.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 06/19/2017] [Indexed: 01/18/2023]
Abstract
Myelodysplastic syndrome (MDS) with deletion 5q (del(5q)) is a distinct clinical and pathological disease subset that is exquisitely sensitive to lenalidomide for the treatment of red blood cell transfusion-dependent anemia. Although lenalidomide has erythropoeitic promoting activity in MDS without del(5q) (non-del(5q) MDS), the frequency of response to treatment is lower and relates to biologically separate drug effects. In del(5q) MDS, lenalidomide suppresses the malignant clone to restore effective erythropoiesis by virtue of synthetic lethality, arising from cereblon-dependent degradation of haplodeficient proteins encoded within the commonly deleted region of the chromosome 5q deletion. In contrast, in non-del(5q) MDS, lenalidomide restores effective erythropoiesis via enhancement of erythropoietin (EPO) receptor-initiated transcriptional response arising from the assembly of signaling-competent receptor complexes within membrane lipid raft domains. Recently, large phase III clinical studies have explored the role of lenalidomide, alone and in combination with, erythropoiesis-stimulating agents showing additive improvement in erythroid responses. Herein, we will describe the mechanisms of lenalidomide action in MDS and pivotal clinical studies testing the benefit of lenalidomide in both del(5q) and non-del(5q) MDS. Furthermore, we discuss evidence-based strategies to incorporate lenalidomide into the treatment algorithm for patients with MDS.
Collapse
Affiliation(s)
- Chetasi Talati
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - David Sallman
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL.
| | - Alan List
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| |
Collapse
|
22
|
Dolatshad H, Pellagatti A, Liberante FG, Llorian M, Repapi E, Steeples V, Roy S, Scifo L, Armstrong RN, Shaw J, Yip BH, Killick S, Kušec R, Taylor S, Mills KI, Savage KI, Smith CWJ, Boultwood J. Cryptic splicing events in the iron transporter ABCB7 and other key target genes in SF3B1-mutant myelodysplastic syndromes. Leukemia 2016; 30:2322-2331. [PMID: 27211273 PMCID: PMC5029572 DOI: 10.1038/leu.2016.149] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/04/2016] [Accepted: 05/16/2016] [Indexed: 02/08/2023]
Abstract
The splicing factor SF3B1 is the most frequently mutated gene in myelodysplastic syndromes (MDS), and is strongly associated with the presence of ring sideroblasts (RS). We have performed a systematic analysis of cryptic splicing abnormalities from RNA sequencing data on hematopoietic stem cells (HSCs) of SF3B1-mutant MDS cases with RS. Aberrant splicing events in many downstream target genes were identified and cryptic 3' splice site usage was a frequent event in SF3B1-mutant MDS. The iron transporter ABCB7 is a well-recognized candidate gene showing marked downregulation in MDS with RS. Our analysis unveiled aberrant ABCB7 splicing, due to usage of an alternative 3' splice site in MDS patient samples, giving rise to a premature termination codon in the ABCB7 mRNA. Treatment of cultured SF3B1-mutant MDS erythroblasts and a CRISPR/Cas9-generated SF3B1-mutant cell line with the nonsense-mediated decay (NMD) inhibitor cycloheximide showed that the aberrantly spliced ABCB7 transcript is targeted by NMD. We describe cryptic splicing events in the HSCs of SF3B1-mutant MDS, and our data support a model in which NMD-induced downregulation of the iron exporter ABCB7 mRNA transcript resulting from aberrant splicing caused by mutant SF3B1 underlies the increased mitochondrial iron accumulation found in MDS patients with RS.
Collapse
Affiliation(s)
- H Dolatshad
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- NIHR Biomedical Research Centre, Oxford, UK
| | - A Pellagatti
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- NIHR Biomedical Research Centre, Oxford, UK
| | - F G Liberante
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - M Llorian
- Department of Biochemistry, Downing Site, University of Cambridge, Cambridge, UK
| | - E Repapi
- The Computational Biology Research Group, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - V Steeples
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- NIHR Biomedical Research Centre, Oxford, UK
| | - S Roy
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- NIHR Biomedical Research Centre, Oxford, UK
| | - L Scifo
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- NIHR Biomedical Research Centre, Oxford, UK
| | - R N Armstrong
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- NIHR Biomedical Research Centre, Oxford, UK
| | - J Shaw
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- NIHR Biomedical Research Centre, Oxford, UK
| | - B H Yip
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- NIHR Biomedical Research Centre, Oxford, UK
| | - S Killick
- Department of Haematology, Royal Bournemouth Hospital, Bournemouth, UK
| | - R Kušec
- Dubrava University hospital and Zagreb School of Medicine, University of Zagreb, Zagreb, Croatia
| | - S Taylor
- The Computational Biology Research Group, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - K I Mills
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - K I Savage
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - C W J Smith
- Department of Biochemistry, Downing Site, University of Cambridge, Cambridge, UK
| | - J Boultwood
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- NIHR Biomedical Research Centre, Oxford, UK
| |
Collapse
|
23
|
Nolte F, Mossner M, Jann JC, Nowak D, Boch T, Müller NZ, Hofmann WK, Metzgeroth G. Concomitant MDS with isolated 5q deletion and MGUS: case report and review of molecular aspects. Eur J Haematol 2016; 98:302-310. [PMID: 27862375 DOI: 10.1111/ejh.12827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2016] [Indexed: 12/23/2022]
Abstract
Patients with monoclonal gammopathy of undetermined significance (MGUS) have a higher risk for the development of concomitant primary cancers such as multiple myeloma (MM) and myelodysplastic syndrome (MDS). We report the case of patient initially suffering from MGUS of the IgG lambda subtype for more than 10 yr, which evolved to MM and MDS with deletion (5q) with severe pancytopenia. Due to pancytopenia, he received dose-reduced treatment with lenalidomide and dexamethasone. He achieved an ongoing transfusion independency after about 1 month of treatment. Bone marrow taken 14 months after start of treatment showed a complete cytogenetic response of the del(5q) clone and a plasma cell infiltration below 5%. In contrast to the development of MM in MGUS patients, the subsequent occurrence of MDS after diagnosis of MGUS is infrequent. Moreover, the biological association of MDS with MGUS is not sufficiently understood, but the non-treatment-related occurrence supports the pathogenetic role of pre-existing alterations of stem cells. Here, we summarize data on concomitant MDS and MGUS/MM with particular emphasis on molecular aspects.
Collapse
Affiliation(s)
- Florian Nolte
- Medical faculty Mannheim of the University of Heidelberg, Mannheim, Germany.,Department of Internal Medicine, Hematology and Oncology, St. Hedwig Hospital, Berlin, Germany
| | - Maximilian Mossner
- Medical faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | | | - Daniel Nowak
- Medical faculty Mannheim of the University of Heidelberg, Mannheim, Germany.,Department of Hematology and Oncology, University Hospital Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Tobias Boch
- Medical faculty Mannheim of the University of Heidelberg, Mannheim, Germany.,Department of Hematology and Oncology, University Hospital Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Nadine Zoe Müller
- Medical faculty Mannheim of the University of Heidelberg, Mannheim, Germany.,Department of Hematology and Oncology, University Hospital Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Wolf-Karsten Hofmann
- Medical faculty Mannheim of the University of Heidelberg, Mannheim, Germany.,Department of Hematology and Oncology, University Hospital Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Georgia Metzgeroth
- Medical faculty Mannheim of the University of Heidelberg, Mannheim, Germany.,Department of Hematology and Oncology, University Hospital Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| |
Collapse
|
24
|
Lian XY, Zhang ZH, Deng ZQ, He PF, Yao DM, Xu ZJ, Wen XM, Yang L, Lin J, Qian J. Efficacy and Safety of Lenalidomide for Treatment of Low-/Intermediate-1-Risk Myelodysplastic Syndromes with or without 5q Deletion: A Systematic Review and Meta-Analysis. PLoS One 2016; 11:e0165948. [PMID: 27824902 PMCID: PMC5100926 DOI: 10.1371/journal.pone.0165948] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 10/20/2016] [Indexed: 01/22/2023] Open
Abstract
Background Lenalidomide could effectively induce red blood cell (RBC) transfusion independence (TI) in patients with lower-risk (Low/Intermediate-1) myelodysplastic syndrome (MDS) with or without 5q deletion. However whether lenalidomide ultimately improves the overall survival (OS) of lower-risk MDS patients and reduces the progression to AML remains controversial. Method A meta-analysis was conducted to examine the efficacy and safety of lenalidomide in the treatment of lower-risk MDS. Efficacy was assessed according to erythroid hematologic response (HI-E), cytogenetic response (CyR), OS and AML progression. Safety was evaluated based on the occurrence rates of grades 3–4 adverse events (AEs). Results Seventeen studies were included consisting of a total of 2160 patients. The analysis indicated that the overall rate of HI-E was 58% with 95% confidence interval (CI) of 43–74%. The pooled estimates for the rates of CyR, complete CyR, and partial CyR were 44% (95% CI 19–68%), 21% (95% CI 13–30%) and 23% (95% CI 15–32%), respectively. The patients with 5q deletion had significantly higher rate of HI-E and CyR than those without 5q deletion (P = 0.002 and 0.001, respectively). The incidences of grades 3–4 neutropenia, thrombocytopenia, leukopenia, anemia, deep vein thrombosis, diarrhea, fatigue and rash were 51% (95% CI 30–73%), 31% (95% CI 20–42%), 9% (95% CI 5–13%), 7% (95% CI 2–12%), 3% (95% CI 2–5%), 3% (95% CI 1–5%), 2% (95% CI 1–4%) and 2% (95% CI 1–3%), respectively. Lenalidomide significantly improved OS (HR: 0.62, 95% CI 0.47–0.83, P = 0.001) and lowered the risk of AML progression in del(5q) patients (RR: 0.61, 95% CI 0.41–0.91, P = 0.014). Conclusions In spite of the AEs, lenalidomide could be effectively and safely used for the treatment of lower-risk MDS patients with or without 5q deletion.
Collapse
Affiliation(s)
- Xin-yue Lian
- Department of Hematology, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Zhi-hui Zhang
- Department of Hematology, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Zhao-qun Deng
- Laboratory Center, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Pin-fang He
- Laboratory Center, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Dong-ming Yao
- Laboratory Center, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Zi-jun Xu
- Laboratory Center, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Xiang-mei Wen
- Laboratory Center, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Lei Yang
- Laboratory Center, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Jiang Lin
- Laboratory Center, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
- * E-mail: (JQ); (JL)
| | - Jun Qian
- Department of Hematology, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
- * E-mail: (JQ); (JL)
| |
Collapse
|
25
|
Fang J, Liu X, Bolanos L, Barker B, Rigolino C, Cortelezzi A, Oliva EN, Cuzzola M, Grimes HL, Fontanillo C, Komurov K, MacBeth K, Starczynowski DT. A calcium- and calpain-dependent pathway determines the response to lenalidomide in myelodysplastic syndromes. Nat Med 2016; 22:727-34. [PMID: 27294874 PMCID: PMC5507589 DOI: 10.1038/nm.4127] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 05/13/2016] [Indexed: 12/15/2022]
Abstract
Despite the high response rates of individuals with myelodysplastic syndrome (MDS) with deletion of chromosome 5q (del(5q)) to treatment with lenalidomide (LEN) and the recent identification of cereblon (CRBN) as the molecular target of LEN, the cellular mechanism by which LEN eliminates MDS clones remains elusive. Here we performed an RNA interference screen to delineate gene regulatory networks that mediate LEN responsiveness in an MDS cell line, MDSL. We identified GPR68, which encodes a G-protein-coupled receptor that has been implicated in calcium metabolism, as the top candidate gene for modulating sensitivity to LEN. LEN induced GPR68 expression via IKAROS family zinc finger 1 (IKZF1), resulting in increased cytosolic calcium levels and activation of a calcium-dependent calpain, CAPN1, which were requisite steps for induction of apoptosis in MDS cells and in acute myeloid leukemia (AML) cells. In contrast, deletion of GPR68 or inhibition of calcium and calpain activation suppressed LEN-induced cytotoxicity. Moreover, expression of calpastatin (CAST), an endogenous CAPN1 inhibitor that is encoded by a gene (CAST) deleted in del(5q) MDS, correlated with LEN responsiveness in patients with del(5q) MDS. Depletion of CAST restored responsiveness of LEN-resistant non-del(5q) MDS cells and AML cells, providing an explanation for the superior responses of patients with del(5q) MDS to LEN treatment. Our study describes a cellular mechanism by which LEN, acting through CRBN and IKZF1, has cytotoxic effects in MDS and AML that depend on a calcium- and calpain-dependent pathway.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Apoptosis/drug effects
- Apoptosis/genetics
- Calcium/metabolism
- Calcium-Binding Proteins/genetics
- Calpain/drug effects
- Calpain/genetics
- Calpain/metabolism
- Cell Line, Tumor
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Regulatory Networks
- Humans
- Ikaros Transcription Factor/drug effects
- Ikaros Transcription Factor/genetics
- Ikaros Transcription Factor/metabolism
- Immunologic Factors/pharmacology
- Lenalidomide
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Myelodysplastic Syndromes/drug therapy
- Myelodysplastic Syndromes/genetics
- Myelodysplastic Syndromes/metabolism
- Peptide Hydrolases/metabolism
- RNA Interference
- Receptors, G-Protein-Coupled/drug effects
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Thalidomide/analogs & derivatives
- Thalidomide/pharmacology
- Ubiquitin-Protein Ligases
Collapse
Affiliation(s)
- Jing Fang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - Xiaona Liu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - Lyndsey Bolanos
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - Brenden Barker
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - Carmela Rigolino
- Bone Marrow Transplant Unit, Azienda Ospedaliera Bianchi Melacrino Morelli, Reggio Calabria, Italy
| | - Agostino Cortelezzi
- Department of Hematology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Esther N Oliva
- Hematology Unit, Azienda Ospedaliera Bianchi Melacrino Morelli, Reggio Calabria, Italy
| | - Maria Cuzzola
- Bone Marrow Transplant Unit, Azienda Ospedaliera Bianchi Melacrino Morelli, Reggio Calabria, Italy
| | - H Leighton Grimes
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | | | - Kakajan Komurov
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - Kyle MacBeth
- Celgene Corporation, San Francisco, California, USA
| | - Daniel T Starczynowski
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
- Department of Cancer Biology, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
26
|
Maffei R, Colaci E, Fiorcari S, Martinelli S, Potenza L, Luppi M, Marasca R. Lenalidomide in chronic lymphocytic leukemia: the present and future in the era of tyrosine kinase inhibitors. Crit Rev Oncol Hematol 2015; 97:291-302. [PMID: 26454471 DOI: 10.1016/j.critrevonc.2015.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 07/21/2015] [Accepted: 09/24/2015] [Indexed: 01/23/2023] Open
Abstract
Lenalidomide is an immunomodulatory agent (IMiD) clinically active in chronic lymphocytic leukemia (CLL), both in heavily pre-treated patients and upfront. Lenalidomide has a unique mechanism of action in CLL. Its efficacy relies on a multifactorial mode-of-action (MOA), comprising a plethora of immunomodulatory actions, the disruption of mutualistic interactions inside CLL microenvironment and direct effects against leukemic cells. In the last few years, a number of new and highly effective drugs appeared in the scenario of CLL therapeutic options, i.e. tyrosine kinase inhibitors (TKIs), showing a good safety profile and impressive clinical response, also in high-risk patients. In this review, we describe the data from clinical studies about lenalidomide efficacy in CLL and we critically dissect the different mechanisms of action of this drug. We point the attention on open issues, including drug dosage and administration schedule, prediction of clinical response to lenalidomide, and combination therapeutic strategies. This overview would be useful to envision a possible role of lenalidomide in the treatment flow-chart of CLL, exploiting its peculiar MOA and also exploring the possible synergetic effect with new drugs.
Collapse
Affiliation(s)
- Rossana Maffei
- Hematology Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisabetta Colaci
- Hematology Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Stefania Fiorcari
- Hematology Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvia Martinelli
- Hematology Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Leonardo Potenza
- Hematology Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Mario Luppi
- Hematology Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberto Marasca
- Hematology Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
27
|
Conte S, Katayama S, Vesterlund L, Karimi M, Dimitriou M, Jansson M, Mortera-Blanco T, Unneberg P, Papaemmanuil E, Sander B, Skoog T, Campbell P, Walfridsson J, Kere J, Hellström-Lindberg E. Aberrant splicing of genes involved in haemoglobin synthesis and impaired terminal erythroid maturation in SF3B1 mutated refractory anaemia with ring sideroblasts. Br J Haematol 2015; 171:478-90. [PMID: 26255870 PMCID: PMC4832260 DOI: 10.1111/bjh.13610] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 06/25/2015] [Indexed: 02/06/2023]
Abstract
Refractory anaemia with ring sideroblasts (RARS) is distinguished by hyperplastic inefficient erythropoiesis, aberrant mitochondrial ferritin accumulation and anaemia. Heterozygous mutations in the spliceosome gene SF3B1 are found in a majority of RARS cases. To explore the link between SF3B1 mutations and anaemia, we studied mutated RARS CD34+ marrow cells with regard to transcriptome sequencing, splice patterns and mutational allele burden during erythroid differentiation. Transcriptome profiling during early erythroid differentiation revealed a marked up‐regulation of genes involved in haemoglobin synthesis and in the oxidative phosphorylation process, and down‐regulation of mitochondrial ABC transporters compared to normal bone marrow. Moreover, mis‐splicing of genes involved in transcription regulation, particularly haemoglobin synthesis, was confirmed, indicating a compromised haemoglobinization during RARS erythropoiesis. In order to define the phase during which erythroid maturation of SF3B1 mutated cells is most affected, we assessed allele burden during erythroid differentiation in vitro and in vivo and found that SF3B1 mutated erythroblasts showed stable expansion until late erythroblast stage but that terminal maturation to reticulocytes was significantly reduced. In conclusion, SF3B1 mutated RARS progenitors display impaired splicing with potential downstream consequences for genes of key importance for haemoglobin synthesis and terminal erythroid differentiation.
Collapse
Affiliation(s)
- Simona Conte
- Karolinska Institutet, Department of Medicine (Huddinge), Centre for Hematology and Regenerative Medicine, Stockholm, Sweden
| | - Shintaro Katayama
- Karolinska Institutet, Department of Biosciences and Nutrition and Center for Innovative Medicine, Stockholm, Sweden
| | - Liselotte Vesterlund
- Karolinska Institutet, Department of Biosciences and Nutrition and Center for Innovative Medicine, Stockholm, Sweden
| | - Mohsen Karimi
- Karolinska Institutet, Department of Medicine (Huddinge), Centre for Hematology and Regenerative Medicine, Stockholm, Sweden
| | - Marios Dimitriou
- Karolinska Institutet, Department of Medicine (Huddinge), Centre for Hematology and Regenerative Medicine, Stockholm, Sweden
| | - Monika Jansson
- Karolinska Institutet, Department of Medicine (Huddinge), Centre for Hematology and Regenerative Medicine, Stockholm, Sweden
| | - Teresa Mortera-Blanco
- Karolinska Institutet, Department of Medicine (Huddinge), Centre for Hematology and Regenerative Medicine, Stockholm, Sweden
| | - Per Unneberg
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm, Sweden
| | - Elli Papaemmanuil
- Cancer Genetics & Genomics, The Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Birgitta Sander
- Karolinska Institutet, Department of Laboratory Medicine, Division of Pathology, Stockholm, Sweden
| | - Tiina Skoog
- Karolinska Institutet, Department of Biosciences and Nutrition and Center for Innovative Medicine, Stockholm, Sweden
| | - Peter Campbell
- Cancer Genetics & Genomics, The Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Julian Walfridsson
- Karolinska Institutet, Department of Medicine (Huddinge), Centre for Hematology and Regenerative Medicine, Stockholm, Sweden
| | - Juha Kere
- Karolinska Institutet, Department of Biosciences and Nutrition and Center for Innovative Medicine, Stockholm, Sweden
| | - Eva Hellström-Lindberg
- Karolinska Institutet, Department of Medicine (Huddinge), Centre for Hematology and Regenerative Medicine, Stockholm, Sweden
| |
Collapse
|
28
|
Nian Q, Zhang Z, Wei C, Kuang X, Wang X, Wang L. Gene expression profiling in myelodysplastic syndrome after SPARC overexpression associated with Ara-C. Oncol Rep 2015; 34:2072-82. [PMID: 26238482 DOI: 10.3892/or.2015.4139] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 06/30/2015] [Indexed: 12/11/2022] Open
Abstract
Secreted protein acidic and rich in cysteine (SPARC) is involved in many biological processes, including erythropoiesis and cell proliferation. However, the role of SPARC in myelodysplastic syndrome (MDS) remains to be elucidated. Pyrimidine analogue cytosine arabinoside (Ara-C) is among the most effective agents used in the treatment of acute leukemia. The aim of the present study was to determine whether the chemotherapeutic activity of Ara-C was enhanced by the overexpression of SPARC. DNA microarray technology and RNA sequencing were employed to examine differential gene expression in the apoptosis signaling pathway after gene change occurred in cells following drug treatment. The results showed that upregulation of the expression of SPARC induced SKM-1 cell death and inhibited proliferation. Additionally, the apoptotic rate of SPARC overexpression combined with Ara-C increased significantly. Transcription factors CPBP and ZNF333 regulated the 69 genes and long non-coding RNA (lncRNA). Moreover, the mRNA and protein expression of apoptosis-related genes in the DNA microarray results were increased. These results suggest that SPARC expression changes with Ara-C, revealing a possible application in the treatment of MDS.
Collapse
Affiliation(s)
- Qing Nian
- Department of Emergency, Children's Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Zhiqiang Zhang
- Department of Emergency, Children's Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Chunmei Wei
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Xingyi Kuang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Xingyong Wang
- Department of Emergency, Children's Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| |
Collapse
|
29
|
Nian Q, Chi J, Xiao Q, Wei C, Costeas P, Yang Z, Liu L, Wang L. SPARC ectopic overexpression inhibits growth and promotes programmed cell death in acute myeloid leukemia transformed from myelodysplastic syndrome cells, alone and in combination with Ara-C treatment. Oncol Rep 2015; 34:1406-14. [PMID: 26165695 DOI: 10.3892/or.2015.4114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 05/12/2015] [Indexed: 11/05/2022] Open
Abstract
Secreted protein acidic and rich in cysteine (SPARC) has a complex and pleiotropic biological role in cell life during disease. The role of SPARC in myelodysplastic syndrome (MDS) is not yet fully understood. In the present study, we investigated the role of SPARC protein overproduction in the proliferation and apoptosis of SKM-1 cells, an acute myeloid leukemia cell line transformed from MDS. SKM-1 cells were infected with the pGC-GV-SPARC vector. The cells were then assessed for proliferation and cell death following treatment with low-dose cytosine arabinoside (Ara‑C). The microarray analysis results revealed that samples from SPARC‑overexpressed cells compared to SPARC protein, in SKM-1 cells led to proliferation inhibition and promoted programmed cell death and these effects were greater when treated with Ara-C. The mRNA and protein expression levels of SPARC were detected by SPARC overexpression in cells treated with Ara-C resulting in a significant upregulation of the mixed lineage kinase domain-like (MLKL) gene expression and five other genes. The results showed that the necrotic signaling pathway may play a role when the two conditions were combined via the upregulation of the MLKL protein. MLKL upregulation in SPARC overexpressed cells treated with Ara-C, indicates necrosis as a possible cell death process for the SKM-1 cells under these stringent conditions.
Collapse
Affiliation(s)
- Qing Nian
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jianxiang Chi
- The Center for the Study of Haematological Malignancies, Nicosia 2032, Cyprus
| | - Qing Xiao
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Chunmei Wei
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Paul Costeas
- The Center for the Study of Haematological Malignancies, Nicosia 2032, Cyprus
| | - Zesong Yang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Lin Liu
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
30
|
Lenalidomide induces ubiquitination and degradation of CK1α in del(5q) MDS. Nature 2015; 523:183-188. [PMID: 26131937 PMCID: PMC4853910 DOI: 10.1038/nature14610] [Citation(s) in RCA: 623] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 06/03/2015] [Indexed: 02/07/2023]
Abstract
Lenalidomide is a highly effective treatment for myelodysplastic syndrome (MDS) with deletion of chromosome 5q (del(5q)). Here, we demonstrate that lenalidomide induces the ubiquitination of casein kinase 1A1 (CK1α) by the E3 ubiquitin ligase CUL4-RBX1-DDB1-CRBN (known as CRL4(CRBN)), resulting in CK1α degradation. CK1α is encoded by a gene within the common deleted region for del(5q) MDS and haploinsufficient expression sensitizes cells to lenalidomide therapy, providing a mechanistic basis for the therapeutic window of lenalidomide in del(5q) MDS. We found that mouse cells are resistant to lenalidomide but that changing a single amino acid in mouse Crbn to the corresponding human residue enables lenalidomide-dependent degradation of CK1α. We further demonstrate that minor side chain modifications in thalidomide and a novel analogue, CC-122, can modulate the spectrum of substrates targeted by CRL4(CRBN). These findings have implications for the clinical activity of lenalidomide and related compounds, and demonstrate the therapeutic potential of novel modulators of E3 ubiquitin ligases.
Collapse
|
31
|
Loiseau C, Ali A, Itzykson R. New therapeutic approaches in myelodysplastic syndromes: Hypomethylating agents and lenalidomide. Exp Hematol 2015; 43:661-72. [PMID: 26123365 DOI: 10.1016/j.exphem.2015.05.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 05/28/2015] [Accepted: 05/29/2015] [Indexed: 01/17/2023]
Abstract
Recent advances in the treatment of myelodysplastic syndromes have come from the use of the hypomethylating agents decitabine and azacitidine as well as the immunomodulatory drug lenalidomide. Their clinical benefit has been demonstrated by randomized phase III clinical trials, mostly in high-risk and del(5q) myelodysplastic syndromes, respectively. Neither drug, however, appears to eradicate myelodysplastic stem cells, and thus they currently do not represent curative options. Here, we review data from both clinical and translational research on those drugs to identify their molecular and cellular mechanisms of action and to delineate paths for improved treatment allocation and further therapeutic advances in myelodysplastic syndromes.
Collapse
Affiliation(s)
- Clémence Loiseau
- Department of Hematology, Saint-Louis Hospital, Assistance Publique, Hopitaux de Paris, Paris Diderot University, Paris, France
| | - Ashfaq Ali
- Institut National de la Santé et de la Recherche Médicale, Saint-Louis Institute, Paris, France
| | - Raphael Itzykson
- Department of Hematology, Saint-Louis Hospital, Assistance Publique, Hopitaux de Paris, Paris Diderot University, Paris, France; Institut National de la Santé et de la Recherche Médicale, Saint-Louis Institute, Paris, France.
| |
Collapse
|
32
|
Recent Advances in the 5q- Syndrome. Mediterr J Hematol Infect Dis 2015; 7:e2015037. [PMID: 26075044 PMCID: PMC4450650 DOI: 10.4084/mjhid.2015.037] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 04/28/2015] [Indexed: 12/12/2022] Open
Abstract
The 5q- syndrome is the most distinct of the myelodysplastic syndromes (MDS) and patients with this disorder have a deletion of chromosome 5q [del(5q)] as the sole karyotypic abnormality. Several genes mapping to the commonly deleted region of the 5q- syndrome have been implicated in disease pathogenesis in recent years. Haploinsufficiency of the ribosomal gene RPS14 has been shown to cause the erythroid defect in the 5q- syndrome. Loss of the microRNA genes miR-145 and miR-146a has been associated with the thrombocytosis observed in 5q- syndrome patients. Haploinsufficiency of CSNK1A1 leads to hematopoietic stem cell expansion in mice and may play a role in the initial clonal expansion in patients with 5q- syndrome. Moreover, a subset of patients harbor mutation of the remaining CSNK1A1 allele. Mouse models of the 5q- syndrome, which recapitulate the key features of the human disease, indicate that a p53-dependent mechanism underlies the pathophysiology of this disorder. Importantly, activation of p53 has been demonstrated in the human 5q- syndrome. Recurrent TP53 mutations have been associated with an increased risk of disease evolution and with decreased response to the drug lenalidomide in del(5q) MDS patients. Potential new therapeutic agents for del(5q) MDS include the translation enhancer L-leucine.
Collapse
|
33
|
Dan C, Chi J, Wang L. Molecular mechanisms of the progression of myelodysplastic syndrome to secondary acute myeloid leukaemia and implication for therapy. Ann Med 2015; 47:209-17. [PMID: 25861829 DOI: 10.3109/07853890.2015.1009156] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Myelodysplastic syndrome (MDS) includes a heterogeneous group of clonal haematological stem cell disorders characterized by dysplasia, cytopenias, ineffective haematopoiesis, and an increased risk of progression to acute myeloid leukaemia (AML), which is also called secondary AML (sAML). Approximately one-third of patients with MDS will progress to sAML within a few months to a few years, and this type of transformation is more common and rapid in patients with high-risk MDS (HR-MDS). However, the precise mechanisms underlying the evolution of MDS to sAML remain unclear. Currently, chemotherapy for sAML has minimal efficacy. The only method of curing patients with sAML is allogeneic haematopoietic stem cell transplantation (Allo-HSCT). Unfortunately, only a few patients are appropriate for transplantation because this disease primarily affects older adult patients. Additionally, compared to de novo AML, sAML is more difficult to cure, and the prognosis is often worse. Therefore, it is important to clarify the molecular mechanisms of the progression of MDS to sAML and to explore the potent drugs for clinical use. This review will highlight several molecular mechanisms of the progression of MDS to sAML and new therapeutic strategies of this disease.
Collapse
Affiliation(s)
- Chunli Dan
- Department of Haematology, The First Affiliated Hospital of Chongqing Medical University , Chongqing , China
| | | | | |
Collapse
|
34
|
Maddocks K, Wei L, Rozewski D, Jiang Y, Zhao Y, Adusumilli M, Pierceall WE, Doykin C, Cardone MH, Jones JA, Flynn J, Andritsos LA, Grever MR, Byrd JC, Johnson AJ, Phelps MA, Blum KA. Reduced occurrence of tumor flare with flavopiridol followed by combined flavopiridol and lenalidomide in patients with relapsed chronic lymphocytic leukemia (CLL). Am J Hematol 2015; 90:327-33. [PMID: 25639448 DOI: 10.1002/ajh.23946] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 01/08/2015] [Indexed: 12/13/2022]
Abstract
Flavopiridol and lenalidomide have activity in refractory CLL without immunosuppression or opportunistic infections seen with other therapies. We hypothesized that flavopiridol treatment could adequately de-bulk disease prior to lenalidomide therapy, decreasing the incidence of tumor flare with higher doses of lenalidomide. In this Phase I study, the maximum tolerated dose was not reached with treatment consisting of flavopiridol 30 mg m(-2) intravenous bolus (IVB) + 30 mg m(-2) continuous intravenous infusion (CIVI) cycle (C) 1 day (D) 1 and 30 mg m(-2) IVB + 50 mg m(-2) CIVI C1 D8,15 and C2-8 D3,10,17 with lenalidomide 15 mg orally daily C2-8 D1-21. There was no unexpected toxicity seen, including no increased tumor lysis, tumor flare (even at higher doses of lenalidomide) or opportunistic infection. Significant clinical activity was demonstrated, with a 51% response rate in this group of heavily pretreated patients. Biomarker testing confirmed association of mitochondrial priming of the BH3 only peptide Puma with response.
Collapse
Affiliation(s)
- Kami Maddocks
- Division of Hematology; Department of Internal Medicine; The Ohio State University, Comprehensive Cancer Center, The Ohio State University, College of Pharmacy, The Ohio State University; Ohio
| | - Lai Wei
- Division of Hematology; Department of Internal Medicine; The Ohio State University, Comprehensive Cancer Center, The Ohio State University, College of Pharmacy, The Ohio State University; Ohio
| | - Darlene Rozewski
- Division of Hematology; Department of Internal Medicine; The Ohio State University, Comprehensive Cancer Center, The Ohio State University, College of Pharmacy, The Ohio State University; Ohio
| | - Yao Jiang
- Division of Hematology; Department of Internal Medicine; The Ohio State University, Comprehensive Cancer Center, The Ohio State University, College of Pharmacy, The Ohio State University; Ohio
| | - Yuan Zhao
- Division of Hematology; Department of Internal Medicine; The Ohio State University, Comprehensive Cancer Center, The Ohio State University, College of Pharmacy, The Ohio State University; Ohio
| | - Mikhil Adusumilli
- Division of Hematology; Department of Internal Medicine; The Ohio State University, Comprehensive Cancer Center, The Ohio State University, College of Pharmacy, The Ohio State University; Ohio
| | - William E. Pierceall
- Division of Hematology; Department of Internal Medicine; The Ohio State University, Comprehensive Cancer Center, The Ohio State University, College of Pharmacy, The Ohio State University; Ohio
| | - Camille Doykin
- Division of Hematology; Department of Internal Medicine; The Ohio State University, Comprehensive Cancer Center, The Ohio State University, College of Pharmacy, The Ohio State University; Ohio
| | - Michael H. Cardone
- Division of Hematology; Department of Internal Medicine; The Ohio State University, Comprehensive Cancer Center, The Ohio State University, College of Pharmacy, The Ohio State University; Ohio
| | - Jeffrey A. Jones
- Division of Hematology; Department of Internal Medicine; The Ohio State University, Comprehensive Cancer Center, The Ohio State University, College of Pharmacy, The Ohio State University; Ohio
| | - Joseph Flynn
- Division of Hematology; Department of Internal Medicine; The Ohio State University, Comprehensive Cancer Center, The Ohio State University, College of Pharmacy, The Ohio State University; Ohio
| | - Leslie A. Andritsos
- Division of Hematology; Department of Internal Medicine; The Ohio State University, Comprehensive Cancer Center, The Ohio State University, College of Pharmacy, The Ohio State University; Ohio
| | - Michael R. Grever
- Division of Hematology; Department of Internal Medicine; The Ohio State University, Comprehensive Cancer Center, The Ohio State University, College of Pharmacy, The Ohio State University; Ohio
| | - John C. Byrd
- Division of Hematology; Department of Internal Medicine; The Ohio State University, Comprehensive Cancer Center, The Ohio State University, College of Pharmacy, The Ohio State University; Ohio
| | - Amy J. Johnson
- Division of Hematology; Department of Internal Medicine; The Ohio State University, Comprehensive Cancer Center, The Ohio State University, College of Pharmacy, The Ohio State University; Ohio
| | - Mitch A. Phelps
- Division of Hematology; Department of Internal Medicine; The Ohio State University, Comprehensive Cancer Center, The Ohio State University, College of Pharmacy, The Ohio State University; Ohio
| | - Kristie A Blum
- Division of Hematology; Department of Internal Medicine; The Ohio State University, Comprehensive Cancer Center, The Ohio State University, College of Pharmacy, The Ohio State University; Ohio
| |
Collapse
|
35
|
Lenalidomide in non-Hodgkin lymphoma: biological perspectives and therapeutic opportunities. Blood 2015; 125:2471-6. [PMID: 25736312 DOI: 10.1182/blood-2014-11-567792] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 02/22/2015] [Indexed: 12/16/2022] Open
Abstract
Lenalidomide is an immunomodulatory drug (IMiD) with activity in lymphoid malignancies occurring primarily through immune modulation (eg, T-cell immune synapse enhancement and NK-cell/T-cell effector augmentation) and antiproliferative effects. Food and Drug Administration-approved for bortezomib-resistant, relapsed/refractory mantle-cell lymphoma, lenalidomide has demonstrated efficacy in several additional lymphoma subtypes. There are many ongoing clinical trials examining the use of lenalidomide alone or in combinatorial therapy. It will be important in these studies to delineate reliable, predictive biomarkers to optimally integrate lenalidomide into lymphoma treatment paradigms.
Collapse
|
36
|
Merkerova MD, Krejcik Z, Belickova M, Hrustincova A, Klema J, Stara E, Zemanova Z, Michalova K, Cermak J, Jonasova A. Genome‐wide mi
RNA
profiling in myelodysplastic syndrome with del(5q) treated with lenalidomide. Eur J Haematol 2014; 95:35-43. [DOI: 10.1111/ejh.12458] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2014] [Indexed: 12/14/2022]
Affiliation(s)
| | - Zdenek Krejcik
- Institute of Hematology and Blood Transfusion Prague Czech Republic
| | - Monika Belickova
- Institute of Hematology and Blood Transfusion Prague Czech Republic
| | | | - Jiri Klema
- Department of Cybernetics Faculty of Electrical Engineering Czech Technical University Prague Czech Republic
| | - Eliška Stara
- Institute of Hematology and Blood Transfusion Prague Czech Republic
| | - Zuzana Zemanova
- Center of Oncocytogenetics General University Hospital and First Faculty of Medicine Charles University Prague Czech Republic
| | - Kyra Michalova
- Institute of Hematology and Blood Transfusion Prague Czech Republic
- Center of Oncocytogenetics General University Hospital and First Faculty of Medicine Charles University Prague Czech Republic
| | - Jaroslav Cermak
- Institute of Hematology and Blood Transfusion Prague Czech Republic
| | - Anna Jonasova
- First Department of Medicine General University Hospital Prague Czech Republic
| |
Collapse
|
37
|
Jonasova A, Bokorova R, Polak J, Vostry M, Kostecka A, Hajkova H, Neuwirtova R, Siskova M, Sponerova D, Cermak J, Mikulenkova D, Cervinek L, Brezinova J, Michalova K, Fuchs O. High level of full-length cereblon mRNA in lower risk myelodysplastic syndrome with isolated 5q deletion is implicated in the efficacy of lenalidomide. Eur J Haematol 2014; 95:27-34. [PMID: 25284710 DOI: 10.1111/ejh.12457] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2014] [Indexed: 01/22/2023]
Abstract
Downregulation of cereblon (CRBN) gene expression is associated with resistance to the immunomodulatory drug lenalidomide and poor survival outcomes in multiple myeloma (MM) patients. However, the importance of CRBN gene expression in patients with myelodysplastic syndrome (MDS) and its impact on lenalidomide therapy are not clear. In this study, we evaluate cereblon expression in mononuclear cells isolated from bone marrow [23 lower risk MDS patients with isolated 5q deletion (5q-), 37 lower risk MDS patients with chromosome 5 without the deletion of long arms (non-5q-), and 24 healthy controls] and from peripheral blood (38 patients with 5q-, 52 non-5q- patients and 25 healthy controls) to gain insight into, firstly, the role of cereblon in lower risk MDS patients with or without 5q deletion and, secondly, into the mechanisms of lenalidomide action. Patients with 5q- lower risk MDS have the highest levels of CRBN mRNA in comparison with both lower risk MDS without the deletion of long arms of chromosome 5 and healthy controls. CRBN gene expression was measured using the quantitative TaqMan real-time PCR. High levels of CRBN mRNA were detected in all lenalidomide responders during the course of therapy. A significant decrease of the CRBN mRNA level during lenalidomide treatment is associated with loss of response to treatment and disease progression. These results suggest that, similar to the treatment of MM, high levels of full-length CRBN mRNA in lower risk 5q- patients are necessary for the efficacy of lenalidomide.
Collapse
Affiliation(s)
- Anna Jonasova
- 1st Medical Clinic, General University Hospital Prague, Prague, Czech Republic
| | - Radka Bokorova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic.,Faculty of Science, Charles University, Prague, Czech Republic
| | - Jaroslav Polak
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Martin Vostry
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Arnost Kostecka
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Hana Hajkova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Radana Neuwirtova
- 1st Medical Clinic, General University Hospital Prague, Prague, Czech Republic
| | - Magda Siskova
- 1st Medical Clinic, General University Hospital Prague, Prague, Czech Republic
| | - Dana Sponerova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Jaroslav Cermak
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Dana Mikulenkova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | | | - Jana Brezinova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Kyra Michalova
- Center of Oncocytogenetics, General University Hospital, Prague, Czech Republic
| | - Ota Fuchs
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| |
Collapse
|
38
|
Lenalidomide induces immunomodulation in chronic lymphocytic leukemia and enhances antitumor immune responses mediated by NK and CD4 T cells. BIOMED RESEARCH INTERNATIONAL 2014; 2014:265840. [PMID: 25313353 PMCID: PMC4182694 DOI: 10.1155/2014/265840] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 07/21/2014] [Accepted: 08/05/2014] [Indexed: 12/11/2022]
Abstract
Lenalidomide is an immunomodulatory drug with therapeutic activity in chronic lymphocytic leukemia (CLL). However, it has pleiotropic effects, and the mechanism of action responsible for its therapeutic activity has not been well defined yet. Herein, we show that lenalidomide treatment does not have an effect on the proliferation of leukemia cells, but it increases the proliferation of B cells from healthy donors. Lenalidomide did not exert a direct effect on the apoptosis of leukemia cells obtained from CLL patients, although it indirectly induced their apoptosis through the activation of nonmalignant immune cells. Thus, lenalidomide markedly increased the proliferation of NK and CD4 T cells. The effect of lenalidomide on NK cells was secondary to the induction of IL-2 production by CD4 T cells. Accordingly, depletion of T cells or blockade of IL-2 activity completely abrogated the proliferation of NK cells. Additionally, lenalidomide enhanced NK and NKT-like cell-mediated natural cytotoxicity against leukemia cells from CLL patients. Lenalidomide also upregulated CD20 expression on leukemia cells and, accordingly, it had a synergistic effect with rituximab on promoting antibody-dependent cell-mediated cytotoxicity against primary leukemia cells. Overall, these observations provide a support for combining lenalidomide with rituximab as a treatment in CLL.
Collapse
|
39
|
Hematopoietic Cell Transplantation for Older Patients with MDS. Mediterr J Hematol Infect Dis 2014; 6:e2014056. [PMID: 25237469 PMCID: PMC4165501 DOI: 10.4084/mjhid.2014.056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 07/27/2014] [Indexed: 12/21/2022] Open
Abstract
The incidence of myeloid malignancies, including myelodysplastic syndromes (MDS) increases with age. While several therapeutic modalities have been developed, for most of these patients the only treatment with curative potential is allogeneic hematopoietic cell transplantation (HCT). The development of reduced/low intensity transplant conditioning regimens allows to successfully transplant patients in their ‘60s and even ‘70s, although comorbidities may determine who does come to transplantation and who does not. Also, as many as half of the patients will develop graft versus host disease (GVHD), even with HLA matched donors, requiring therapy for extended periods of time, and GVHD and treatment with glucocorticoids is likely to impact the quality of life. Nevertheless, dependent upon disease stage at HCT, the presence of comorbidities and the regimen used, 30% to 50% of patients 60 years of age or older, may survive long-term cured of their disease. Future studies should focus on the incorporation of non-transplant modalities into the overall transplant approach, the prevention of GVHD, and the utilization of immunotherapy to reduce the incidence of relapse and GVHD and further improve overall transplant success.
Collapse
|
40
|
Nakhoul H, Ke J, Zhou X, Liao W, Zeng SX, Lu H. Ribosomopathies: mechanisms of disease. PLASMATOLOGY 2014; 7:7-16. [PMID: 25512719 PMCID: PMC4251057 DOI: 10.4137/cmbd.s16952] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 07/03/2014] [Accepted: 07/16/2014] [Indexed: 01/05/2023]
Abstract
Ribosomopathies are diseases caused by alterations in the structure or function of ribosomal components. Progress in our understanding of the role of the ribosome in translational and transcriptional regulation has clarified the mechanisms of the ribosomopathies and the relationship between ribosomal dysfunction and other diseases, especially cancer. This review aims to discuss these topics with updated information.
Collapse
Affiliation(s)
- Hani Nakhoul
- Department of Biochemistry and Molecular Biology and Cancer Center, Tulane University, School of Medicine, New Orleans, Louisiana, LA, USA
| | - Jiangwei Ke
- Department of Biochemistry and Molecular Biology and Cancer Center, Tulane University, School of Medicine, New Orleans, Louisiana, LA, USA. ; Department of Laboratory Medicine, Jiangxi Children's Hospital, Nanchang, Jiangxi, China
| | - Xiang Zhou
- Department of Biochemistry and Molecular Biology and Cancer Center, Tulane University, School of Medicine, New Orleans, Louisiana, LA, USA
| | - Wenjuan Liao
- Department of Biochemistry and Molecular Biology and Cancer Center, Tulane University, School of Medicine, New Orleans, Louisiana, LA, USA
| | - Shelya X Zeng
- Department of Biochemistry and Molecular Biology and Cancer Center, Tulane University, School of Medicine, New Orleans, Louisiana, LA, USA
| | - Hua Lu
- Department of Biochemistry and Molecular Biology and Cancer Center, Tulane University, School of Medicine, New Orleans, Louisiana, LA, USA
| |
Collapse
|
41
|
Maddocks K, Ruppert AS, Browning R, Jones J, Flynn J, Kefauver C, Gao Y, Jiang Y, Rozewski DM, Poi M, Phelps MA, Harper E, Johnson AJ, Byrd JC, Andritsos LA. A dose escalation feasibility study of lenalidomide for treatment of symptomatic, relapsed chronic lymphocytic leukemia. Leuk Res 2014; 38:1025-9. [PMID: 25082342 DOI: 10.1016/j.leukres.2014.05.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 04/13/2014] [Accepted: 05/18/2014] [Indexed: 11/19/2022]
Abstract
Adequate dosing of lenalidomide in Chronic Lymphocytic Leukemia (CLL) remains unclear. This study determined maximum tolerated dose (MTD) in relapsed CLL patients (Cohort A) and patients achieving a partial response (PR) or better to recent therapy (Cohort B). Thirty-seven patients were enrolled. MTD was 2.5mg followed by 5.0mg continuous. In Cohort A, tumor flare grade 1-2 occurred in 15 patients (50%) and grade 3 in 1 patient (3%). Cohort A had 19 of 23 evaluable (83%) patients, 4 PR (17%) and 15 (65%) stable disease (SD), Cohort B had 6 of 7 patients (86%) with SD. Despite overall response rate not being high, many patients remained on therapy several months with SD.
Collapse
Affiliation(s)
- Kami Maddocks
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA; Comprehensive Cancer Center, The Ohio State University, USA.
| | - Amy S Ruppert
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | | | - Jeffrey Jones
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA; Comprehensive Cancer Center, The Ohio State University, USA
| | - Joseph Flynn
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA; Comprehensive Cancer Center, The Ohio State University, USA
| | | | - Yue Gao
- Division of Pharmaceutics, College of Pharmacy, USA
| | - Yao Jiang
- Division of Pharmaceutics, College of Pharmacy, USA
| | | | - Ming Poi
- Division of Pharmaceutics, College of Pharmacy, USA
| | - Mitch A Phelps
- Comprehensive Cancer Center, The Ohio State University, USA; Division of Pharmaceutics, College of Pharmacy, USA
| | - Erica Harper
- Comprehensive Cancer Center, The Ohio State University, USA
| | - Amy J Johnson
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA; Comprehensive Cancer Center, The Ohio State University, USA
| | - John C Byrd
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA; Comprehensive Cancer Center, The Ohio State University, USA
| | - Leslie A Andritsos
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA; Comprehensive Cancer Center, The Ohio State University, USA
| |
Collapse
|
42
|
Loss of SPARC protects hematopoietic stem cells from chemotherapy toxicity by accelerating their return to quiescence. Blood 2014; 123:4054-63. [PMID: 24833352 DOI: 10.1182/blood-2013-10-533711] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Around birth, hematopoietic stem cells (HSCs) expanding in the fetal liver migrate to the developing bone marrow (BM) to mature and expand. To identify the molecular processes associated with HSCs located in the 2 different microenvironments, we compared the expression profiles of HSCs present in the liver and BM of perinatal mice. This revealed the higher expression of a cluster of extracellular matrix-related genes in BM HSCs, with secreted protein acidic and rich in cysteine (SPARC) being one of the most significant ones. This extracellular matrix protein has been described to be involved in tissue development, repair, and remodeling, as well as metastasis formation. Here we demonstrate that SPARC-deficient mice display higher resistance to serial treatment with the chemotherapeutic agent 5-fluorouracil (5-FU). Using straight and reverse chimeras, we further show that this protective effect is not due to a role of SPARC in HSCs, but rather is due to its function in the BM niche. Although the kinetics of recovery of the hematopoietic system is normal, HSCs in a SPARC-deficient niche show an accelerated return to quiescence, protecting them from the lethal effects of serial 5-FU treatment. This may become clinically relevant, as SPARC inhibition and its protective effect on HSCs could be used to optimize chemotherapy schemes.
Collapse
|
43
|
Gaballa MR, Besa EC. Myelodysplastic syndromes with 5q deletion: pathophysiology and role of lenalidomide. Ann Hematol 2014; 93:723-33. [PMID: 24627193 DOI: 10.1007/s00277-014-2022-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 01/24/2014] [Indexed: 12/19/2022]
Abstract
Myelodysplastic syndrome (MDS) is a hematopoietic stem cell disorder primarily affecting CD34+ cells, characterized by ineffective hematopoiesis, often transforming into acute myelogenous leukemia (AML). A subset of patients has 5q deletion (del(5q)) as the culprit pathogenetic trigger. Del(5q) affects critical regions 5q31 and 5q33, leading to gene haplodeficiency with subsequent RPS14 haplodeficiency and P53 activation. Subsequent to P53 activation, erythroid cell apoptosis and ineffective erythropoiesis occur. Other pathogenetic elements include protein phosphatase 2a and CDC25C haplodeficiency and decreased miR-145 and miR-146a expression. Lenalidomide is an immunomodulatory agent that selectively suppresses the del(5q) clone. While the mechanism is not fully understood, it is associated with diverse molecular changes including stabilization of MDM2 with subsequent enhanced P53 degradation. Lenalidomide showed success in low- and intermediate-1-risk MDS as reported in the 002, 003, and 004 trials. However, in higher-risk MDS, the results of lenalidomide monotherapy were modest, mandating the use of combination therapy. The role and priority of lenalidomide varies between different guidelines, and accordingly, future efforts are necessary to reach a unified therapeutic algorithm. TP53 mutations are important predictors of AML progression and possible resistance to lenalidomide. It is recommended to identify TP53 mutation early in the disease since it may change the decision regarding choice of therapy. Challenges with lenalidomide therapy remain the long-term effects and timing of its discontinuation.
Collapse
Affiliation(s)
- Mahmoud R Gaballa
- Department of Internal Medicine, Thomas Jefferson University, 833 Chestnut Street, Suit 701, Philadelphia, PA, 19107, USA,
| | | |
Collapse
|
44
|
Alachkar H, Santhanam R, Maharry K, Metzeler KH, Huang X, Kohlschmidt J, Mendler JH, Benito JM, Hickey C, Neviani P, Dorrance AM, Anghelina M, Khalife J, Tarighat SS, Volinia S, Whitman SP, Paschka P, Hoellerbauer P, Wu YZ, Han L, Bolon BN, Blum W, Mrózek K, Carroll AJ, Perrotti D, Andreeff M, Caligiuri MA, Konopleva M, Garzon R, Bloomfield CD, Marcucci G. SPARC promotes leukemic cell growth and predicts acute myeloid leukemia outcome. J Clin Invest 2014; 124:1512-24. [PMID: 24590286 DOI: 10.1172/jci70921] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 01/02/2014] [Indexed: 12/20/2022] Open
Abstract
Aberrant expression of the secreted protein, acidic, cysteine-rich (osteonectin) (SPARC) gene, which encodes a matricellular protein that participates in normal tissue remodeling, is associated with a variety of diseases including cancer, but the contribution of SPARC to malignant growth remains controversial. We previously reported that SPARC was among the most upregulated genes in cytogenetically normal acute myeloid leukemia (CN-AML) patients with gene-expression profiles predictive of unfavorable outcome, such as mutations in isocitrate dehydrogenase 2 (IDH2-R172) and overexpression of the oncogenes brain and acute leukemia, cytoplasmic (BAALC) and v-ets erythroblastosis virus E26 oncogene homolog (ERG). In contrast, SPARC was downregulated in CN-AML patients harboring mutations in nucleophosmin (NPM1) that are associated with favorable prognosis. Based on these observations, we hypothesized that SPARC expression is clinically relevant in AML. Here, we found that SPARC overexpression is associated with adverse outcome in CN-AML patients and promotes aggressive leukemia growth in murine models of AML. In leukemia cells, SPARC expression was mediated by the SP1/NF-κB transactivation complex. Furthermore, secreted SPARC activated the integrin-linked kinase/AKT (ILK/AKT) pathway, likely via integrin interaction, and subsequent β-catenin signaling, which is involved in leukemia cell self-renewal. Pharmacologic inhibition of the SP1/NF-κB complex resulted in SPARC downregulation and leukemia growth inhibition. Together, our data indicate that evaluation of SPARC expression has prognosticative value and SPARC is a potential therapeutic target for AML.
Collapse
|
45
|
Heise C, Carter T, Schafer P, Chopra R. Pleiotropic mechanisms of action of lenalidomide efficacy in del(5q) myelodysplastic syndromes. Expert Rev Anticancer Ther 2014; 10:1663-72. [DOI: 10.1586/era.10.135] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
46
|
Clave E, Douay C, Coman T, Busson M, Bompoint C, Moins-Teisserenc H, Glauzy S, Carmagnat M, Gorin NC, Toubert A, Garderet L. Lenalidomide consolidation and maintenance therapy after autologous stem cell transplant for multiple myeloma induces persistent changes in T-cell homeostasis. Leuk Lymphoma 2013; 55:1788-95. [PMID: 24237448 DOI: 10.3109/10428194.2013.865182] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Whether the efficacy of lenalidomide in the treatment of multiple myeloma (MM) is due to direct tumor toxicity only or to additional immunomodulatory effects is unclear. We studied the effect of lenalidomide treatment on T-cell immune reconstitution in patients with MM who had undergone autologous peripheral blood stem cell transplant (ASCT). Twenty-nine newly diagnosed patients with MM received induction therapy followed by high-dose melphalan and ASCT. After ASCT, 11 patients received lenalidomide consolidation therapy for 2 months followed by maintenance therapy until disease progression. The remaining 18 patients received no treatment. Serial analysis of thymic output, as given by numbers of T-cell receptor excision circles (sjTRECs), and T-cell phenotyping was performed until 18 months post-ASCT. Lenalidomide impaired long-term thymic T-cell reconstitution, decreased CD4 + and CD8 + CD45RA + CCR7 - effector-terminal T-cell absolute counts and increased CD4 + CD25 + CD127 - /low regulatory T-cells. Lenalidomide consolidation and long-term maintenance therapy, administered post-ASCT, may have a potentially negative impact on immune surveillance.
Collapse
Affiliation(s)
- Emmanuel Clave
- INSERM UMRS 940, AP-HP, Hôpital Saint Louis, Département d'immunologie, Université Paris Diderot-Paris 7 , Paris , France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Song K, Herzog BH, Sheng M, Fu J, McDaniel JM, Chen H, Ruan J, Xia L. Lenalidomide inhibits lymphangiogenesis in preclinical models of mantle cell lymphoma. Cancer Res 2013; 73:7254-64. [PMID: 24158094 DOI: 10.1158/0008-5472.can-13-0750] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Lymphomas originate in and spread primarily along the lymphatic system. However, whether lymphatic vessels contribute to the growth and spreading of lymphomas is largely unclear. Mantle cell lymphoma (MCL) represents an aggressive non-Hodgkin's lymphoma. We found that MCL exhibited abundant intratumor lymphatic vessels. Our results demonstrated that the immunomodulatory drug lenalidomide potently inhibited the growth and dissemination of MCL in a xenograft MCL mouse model, at least in part, by inhibiting functional tumor lymphangiogenesis. Significant numbers of tumor-associated macrophages expressing vascular endothelial growth factor-C were found in both human MCL and mouse MCL xenograft samples. Lenalidomide treatment resulted in a significant reduction in the number of MCL-associated macrophages. In addition, in vivo depletion of monocytes/macrophages impaired functional tumor lymphangiogenesis and inhibited MCL growth and dissemination. Taken together, our results indicate that tumor lymphangiogenesis contributes to the progression of MCL and that lenalidomide is effective in decreasing MCL growth and metastasis most likely by inhibiting recruitment of MCL-associated macrophages.
Collapse
Affiliation(s)
- Kai Song
- Authors' Affiliations: Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation; Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Center for Lymphoma and Myeloma, Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York City, New York; China-Japan Union Hospital of Jilin University, Changchun; Key Laboratory of Thrombosis and Hemostasis of Ministry of Health; and Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Faltas B, Zeidan A, Gergis U. Myelodysplastic syndromes: toward a risk-adapted treatment approach. Expert Rev Hematol 2013; 6:611-24. [PMID: 24094045 DOI: 10.1586/17474086.2013.840997] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Several classification and scoring systems have been developed in myelodysplastic syndromes (MDS to predict the risk of progression to acute myeloid leukemia and survival. These prognostication models have been also used to inform therapeutic decision-making in a risk-adapted fashion. Patient-related factors such as age, comorbidities, and functional status have to be considered as well. Here we review a risk-guided therapeutic approach for the management of MDS patients. It is anticipated that the improved understanding of the complex pathogenesis of MDS and the recent discovery of important molecular lesions will be translated into novel therapeutic approaches. Additionally, some prognostic aberrations are expected to be incorporated into the prognostic tools with the goal of improving their prognostic precision and therefore allow for a more informed therapeutic decision-making based on the individual's risk profile.
Collapse
Affiliation(s)
- Bishoy Faltas
- Division of Hematology and Medical Oncology, Weill-Medical College of Cornell University/New York Presbyterian Hospital, NY 10065, USA
| | | | | |
Collapse
|
49
|
Boultwood J, Dolatshad H, Varanasi SS, Yip BH, Pellagatti A. The role of splicing factor mutations in the pathogenesis of the myelodysplastic syndromes. Adv Biol Regul 2013; 54:153-61. [PMID: 24080589 DOI: 10.1016/j.jbior.2013.09.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 09/03/2013] [Accepted: 09/03/2013] [Indexed: 11/26/2022]
Abstract
Accurate pre-mRNA splicing by the spliceosome is a fundamental cellular mechanism required to remove introns that are present in most protein-coding transcripts. The recent discovery of a variety of somatic spliceosomal mutations in the myelodysplastic syndromes (MDS), a heterogeneous group of myeloid malignancies, has revealed a new leukemogenic pathway involving spliceosomal dysfunction. Spliceosome mutations are found in over half of all MDS patients and are likely founder mutations. The spliceosome mutations are highly specific to MDS and closely related conditions and, to some extent, appear to define distinct clinical phenotypes in MDS. The high frequency of mutations in different components of the RNA splicing machinery in MDS suggests that abnormal RNA splicing is the common consequence of these mutations. The identification of the downstream targets of the spliceosome mutations is an active area of research. Emerging data from the study of the MDS transcriptome suggests that spliceosomal mutations have effects on specific genes, including some previously shown to play a role in MDS pathogenesis. The effects of the spliceosomal mutations on RNA splicing and cell growth have been evaluated only in a limited context to date, however, and the determination of the impact of these mutations in primary human hematopoietic cells is essential in order to elucidate fully the molecular mechanism by which they contribute to MDS pathogenesis.
Collapse
Affiliation(s)
- Jacqueline Boultwood
- LLR Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom.
| | - Hamid Dolatshad
- LLR Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom
| | - Satya S Varanasi
- LLR Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom
| | - Bon Ham Yip
- LLR Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom
| | - Andrea Pellagatti
- LLR Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom
| |
Collapse
|
50
|
Giagounidis A, Mufti GJ, Fenaux P, Germing U, List A, MacBeth KJ. Lenalidomide as a disease-modifying agent in patients with del(5q) myelodysplastic syndromes: linking mechanism of action to clinical outcomes. Ann Hematol 2013; 93:1-11. [PMID: 24018623 PMCID: PMC3889654 DOI: 10.1007/s00277-013-1863-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Accepted: 07/23/2013] [Indexed: 12/01/2022]
Abstract
Deletion of the long arm of chromosome 5, del(5q), is the most prevalent cytogenetic abnormality in patients with myelodysplastic syndromes (MDS). In isolation, it is traditionally associated with favorable prognosis compared with other subtypes of MDS. However, owing to the inherent heterogeneity of the disease, prognosis for patients with del(5q) MDS is highly variable depending on the presence of factors such as additional chromosomal abnormalities, >5 % blasts in the bone marrow (BM), or transfusion dependence. Over recent years, the immunomodulatory drug lenalidomide has demonstrated remarkable efficacy in patients with del(5q) MDS. Advances in the understanding of the pathogenesis of the disease have suggested that lenalidomide targets aberrant signaling pathways caused by haplosufficiency of specific genes in a commonly deleted region on chromosome 5 (e.g., SPARC, RPS14, Cdc25C, and PP2A). As a result, the agent specifically targets del(5q) clones while also promoting erythropoiesis and repopulation of the bone marrow in normal cells. This review discusses recent developments in the understanding of the mechanism of action of lenalidomide, and how this underlies favorable outcomes in patients with del(5q) MDS. In addition, we discuss how improved understanding of the mechanism of disease will facilitate clinicians’ ability to predict/monitor response and identify patients at risk of relapse.
Collapse
|