1
|
SUKU PRATIBHA, DASH AISHWARYA, RADHAKRISHNAN ARAVIND, MALHOTRA PANKAJ, SACHDEVA MANUPDESHSINGH. A review on pathobiology of circulating tumour plasma cells: The sine qua non of poor prognosis in plasma cell neoplasms. Oncol Res 2025; 33:1055-1068. [PMID: 40296907 PMCID: PMC12034004 DOI: 10.32604/or.2024.055154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 12/13/2024] [Indexed: 04/30/2025] Open
Abstract
Circulating plasma cells (CPCs) in patients of plasma cell neoplasm have been an area of intense research in recent decades. Circulating tumor plasma cells (CTPCs) might represent a sub-clone of tumor cells that have exited into peripheral blood as a result of the dynamic interactions between the bone marrow (BM) microenvironment and neoplastic plasma cells. Chemokine receptors like chemokine receptor 4 (CXCR4) and integrins are known to play a role in homing and migration of plasma cells (PCs). The hypoxic microenvironment in the BM niche also contributes to their circulation through various mechanisms. In addition, the CCL3-CCR1 axis probably competes with the retention signals from the CXCR4-α4β1 (VLA-4) interaction and actively promotes the exit of PCs from the BM. CTPCs, even in extremely low numbers, can be detected and quantified by high-sensitivity techniques like multi-color flow cytometry and next-generation sequencing. High load of CTPCs noted in patients of plasma cell neoplasm; monoclonal gammopathy of undetermined significance (MGUS), smoldering multiple myeloma (SMM), multiple myeloma (MM) is a strong predictor of shorter progression free survival (PFS) as well as overall survival (OS). In newly diagnosed patients of MM, a load of CTPCs correlates with the outcomes, i.e., OS and PFS. With more studies collaborating on the results of previous reports, assessment of the burden of CTPCs may become a complimentary approach for non-invasive risk stratification of MM patients and evaluating the response to therapy. Future research on larger cohorts and longer follow-ups may help to improve the existing staging system by incorporating the load of CTPCs as one of the prognostic indicators. Further studies based on isolation and genetic characterization of CTPCs may help in understanding the pathophysiology of the progression of the disease and may open avenues for newer treatment modalities. This review discusses the pathobiological aspects leading to circulation of neoplastic/tumor plasma cells in peripheral blood and provides a summary of research work done in last two decades on its prognostic importance in various plasma cells neoplasms.
Collapse
Affiliation(s)
- PRATIBHA SUKU
- Department of Hematology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, P.O. Box 160012, India
| | - AISHWARYA DASH
- Department of Hematology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, P.O. Box 160012, India
| | - ARAVIND RADHAKRISHNAN
- Department of Hematology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, P.O. Box 160012, India
| | - PANKAJ MALHOTRA
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, P.O. Box 160012, India
| | - MAN UPDESH SINGH SACHDEVA
- Department of Hematology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, P.O. Box 160012, India
| |
Collapse
|
2
|
McAuley N, Cymer I, McAvera R, Hopkins AM, Glavey SV. Chromosome 1 Alterations in Multiple Myeloma: Considerations for Precision Therapy. Eur J Haematol 2025; 114:400-410. [PMID: 39632279 PMCID: PMC11798765 DOI: 10.1111/ejh.14352] [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: 09/25/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024]
Abstract
Multiple myeloma (MM) is an incurable blood malignancy characterized by the clonal expansion of plasma cells and the secretion of monoclonal immunoglobulins. High-risk MM, defined by specific cytogenetic abnormalities, poses significant therapeutic challenges and is associated with inferior survival outcomes compared to standard-risk disease. Although molecularly targeted therapies have shown efficacy in other hematologic malignancies, currently venetoclax is the only targeted therapy approved for MM (t(11;14)). However, chromosome 1q gains, amplifications, and 1p deletions are frequently observed in MM, and have been linked to drug resistance and poor patient prognosis. Accordingly, this review focuses on emerging MM precision therapies capable of targeting dysregulated genes within these regions. It addresses gene therapies, small molecule inhibitors and monoclonal antibodies currently under investigation to antagonize oncogenic drivers including MCL-1, BCL9, F11R, and CKS1B, all of which are implicated in cell survival, proliferation or drug resistance. In conclusion, the link between chromosome 1 abnormalities and high-risk disease in MM patients offers a compelling rationale to identify and explore therapeutic targeting of chromosome 1 gene products as a novel precision medicine approach for a poorly served patient population.
Collapse
Affiliation(s)
- Niamh McAuley
- Department of PathologyRCSI University of Medicine and Health SciencesDublinIreland
- Department of SurgeryRCSI University of Medicine and Health SciencesDublinIreland
| | - Izabela Cymer
- Department of PathologyRCSI University of Medicine and Health SciencesDublinIreland
- Department of SurgeryRCSI University of Medicine and Health SciencesDublinIreland
| | - Roisin McAvera
- Department of PathologyRCSI University of Medicine and Health SciencesDublinIreland
- Department of SurgeryRCSI University of Medicine and Health SciencesDublinIreland
| | - Ann M. Hopkins
- Department of SurgeryRCSI University of Medicine and Health SciencesDublinIreland
| | - Siobhan V. Glavey
- Department of PathologyRCSI University of Medicine and Health SciencesDublinIreland
- Department of HaematologyBeaumont RCSI Cancer CentreDublinIreland
| |
Collapse
|
3
|
Lema Fernandez AG, Nardelli C, Pierini V, Crescenzi B, Pellanera F, Matteucci C, Crocioni M, Arniani S, Di Battista V, Quintini M, Mondanelli G, Orabona C, Gorello P, Mecucci C. Epigenetic Modeling of Jumping Translocations of 1q Heterochromatin in Acute Myeloid Leukemia After 5'-Azacytidine Treatment. Genes Chromosomes Cancer 2024; 63:e70013. [PMID: 39604137 PMCID: PMC11602642 DOI: 10.1002/gcc.70013] [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: 09/05/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Jumping translocations (JT) are rare cytogenetic abnormalities associated with progression in myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Typically, a tri-tetra-somic 1q chromosome is translocated to two or more recipient chromosomes. In multiple myeloma JT were shown to originate after DNA demethylation and decondensation. Using epigenomics, we investigated sequential samples in an SRSF2-mutated MDS and AML cohort with normal karyotype at diagnosis and 1qJT at disease evolution after 5'-azacytidine (AZA). 1qJT breakpoints fell within repetitive DNA at both 1q12 and the translocation partners, namely acrocentrics n. 14, 15, 21, and 22, chromosome 16, and chromosome Y. The global methylome at diagnosis showed hypermethylation at 61% of the differentially methylated regions (DMRs), followed by hypomethylation at 80% of DMRs under AZA, mostly affecting pathways related to immune system, chromatin organization, chromosome condensation, telomere maintenance, rRNA, and DNA repair. At disease evolution, a shift toward hypermethylation, intronic enhancers enrichment and epigenetic involvement of the PI3K/AKT and MAPK signaling emerged. In particular, AKT1 phosphorylation behaved as a hallmark of the progression. Overall, we provided new insights on the characterization of 1qJT in SRSF2-mutated myeloid neoplasms and first showed that epigenetics is a powerful tool to investigate the molecular landscape of repetitive DNA rearrangements.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Translocation, Genetic
- Epigenesis, Genetic
- Heterochromatin/genetics
- Chromosomes, Human, Pair 1/genetics
- Azacitidine/pharmacology
- DNA Methylation
- Female
- Male
- Aged
- Middle Aged
- Myelodysplastic Syndromes/genetics
- Myelodysplastic Syndromes/drug therapy
- Myelodysplastic Syndromes/pathology
Collapse
Affiliation(s)
- Anair Graciela Lema Fernandez
- Department of Medicine and Surgery, Hematology and Bone Marrow Transplantation UnitUniversity of PerugiaPerugiaItaly
| | - Carlotta Nardelli
- Department of Medicine and Surgery, Hematology and Bone Marrow Transplantation UnitUniversity of PerugiaPerugiaItaly
| | - Valentina Pierini
- Department of Medicine and Surgery, Hematology and Bone Marrow Transplantation UnitUniversity of PerugiaPerugiaItaly
| | - Barbara Crescenzi
- Department of Medicine and Surgery, Hematology and Bone Marrow Transplantation UnitUniversity of PerugiaPerugiaItaly
| | - Fabrizia Pellanera
- Department of Medicine and Surgery, Hematology and Bone Marrow Transplantation UnitUniversity of PerugiaPerugiaItaly
| | - Caterina Matteucci
- Department of Medicine and Surgery, Hematology and Bone Marrow Transplantation UnitUniversity of PerugiaPerugiaItaly
| | - Maria Crocioni
- Department of Medicine and Surgery, Hematology and Bone Marrow Transplantation UnitUniversity of PerugiaPerugiaItaly
| | - Silvia Arniani
- Department of Medicine and Surgery, Hematology and Bone Marrow Transplantation UnitUniversity of PerugiaPerugiaItaly
| | - Valeria Di Battista
- Department of Medicine and Surgery, Hematology and Bone Marrow Transplantation UnitUniversity of PerugiaPerugiaItaly
| | - Martina Quintini
- Department of Medicine and Surgery, Hematology and Bone Marrow Transplantation UnitUniversity of PerugiaPerugiaItaly
| | - Giada Mondanelli
- Department of Medicine and Surgery, Section of PharmacologyUniversity of PerugiaPerugiaItaly
| | - Ciriana Orabona
- Department of Medicine and Surgery, Section of PharmacologyUniversity of PerugiaPerugiaItaly
| | - Paolo Gorello
- Department of Medicine and Surgery, Hematology and Bone Marrow Transplantation UnitUniversity of PerugiaPerugiaItaly
- Department of Chemistry, Biology and BiotechnologyUniversity of PerugiaPerugiaItaly
| | - Cristina Mecucci
- Department of Medicine and Surgery, Hematology and Bone Marrow Transplantation UnitUniversity of PerugiaPerugiaItaly
| |
Collapse
|
4
|
Liu L, Gong D, Sun H, Feng F, Xu J, Sun X, Gong L, Yu Z, Fang T, Xu Y, Lyu R, Wang T, Wang W, Tian W, Qiu L, An G, Hao M. DNp73 enhances tumor progression and immune evasion in multiple myeloma by targeting the MYC and MYCN pathways. Front Immunol 2024; 15:1470328. [PMID: 39380995 PMCID: PMC11459316 DOI: 10.3389/fimmu.2024.1470328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/04/2024] [Indexed: 10/10/2024] Open
Abstract
Introduction Multiple myeloma (MM) is an incurable hematological malignancy with high chromosome instability and heavy dependence on the immunosuppressive bone marrow microenvironment. P53 mutations are adverse prognostic factors in MM; however, clinically, some patients without P53 mutations also exhibit aggressive disease progression. DNp73, an inhibitor of TP53 tumor suppressor family members, drives drug resistance and cancer progression in several solid malignancies. Nevertheless, the biological functions of DNp73 and the molecular mechanisms in myelomagenesis remain unclear. Methods The effects of DNp73 on proliferation and drug sensitivity were assessed using flow cytometry and xenograft models. To investigate the mechanisms of drug resistance, RNA-seq and ChIP-seq analyses were performed in MM cell lines, with validation by Western blot and RT-qPCR. Immunofluorescence and transwell assays were used to assess DNA damage and cell invasion in MM cells. Additionally, in vitro phagocytosis assays were conducted to confirm the role of DNp73 in immune evasion. Results Our study found that activation of NF-κB-p65 in multiple myeloma cells with different p53 mutation statuses upregulates DNp73 expression at the transcriptional level. Forced expression of DNp73 promoted aggressive proliferation and multidrug resistance in MM cells. Bulk RNA-seq analysis was conducted to assess the levels of MYCN, MYC, and CDK7. A ChIP-qPCR assay was used to reveal that DNp73 acts as a transcription factor regulating MYCN gene expression. Bulk RNA-seq analysis demonstrated increased levels of MYCN, MYC, and CDK7 with forced DNp73 expression in MM cells. A ChIP-qPCR assay revealed that DNp73 upregulates MYCN gene expression as a transcription factor. Additionally, DNp73 promoted immune evasion of MM cells by upregulating MYC target genes CD47 and PD-L1. Blockade of the CD47/SIRPα and PD-1/PD-L1 signaling pathways by the SIRPα-Fc fusion protein IMM01 and monoclonal antibody atezolizumab significantly restored the anti-MM activity of macrophages and T cells in the microenvironment, respectively. Discussion In summary, our study demonstrated for the first time that the p53 family member DNp73 remarkably induces proliferation, drug resistance, and immune escape of myeloma cells by directly targeting MYCN and regulating the MYC pathway. The oncogenic function of DNp73 is independent of p53 status in MM cells. These data contribute to a better understanding of the function of TP53 and its family members in tumorigenesis. Moreover, our study clarified that DNp73 overexpression not only promotes aggressive growth of tumor cells but, more importantly, promotes immune escape of MM cells through upregulation of immune checkpoints. DNp73 could serve as a biomarker for immunotherapy targeting PD-L1 and CD47 blockade in MM patients.
Collapse
Affiliation(s)
- Lanting Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Dasen Gong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Tianjin, China
| | - Hao Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Fangshuo Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jie Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Xiyue Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lixin Gong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Zhen Yu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Teng Fang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yan Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Rui Lyu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Tingyu Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wentian Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wenzhi Tian
- ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, China
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
- Gobroad Healthcare Group, Beijing, China
| | - Gang An
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Mu Hao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
5
|
Halper-Stromberg E, Stinnett V, Morsberger L, Pallavajjala A, Levis MJ, DeZern AE, Lei M, Phan B, Xian RR, Gocke CD, Tang G, Zou YS. 1q jumping translocation as a biomarker in myeloid malignancy: frequently mutated genes associated with bad prognosis and low survival. Exp Hematol Oncol 2024; 13:73. [PMID: 39090734 PMCID: PMC11295323 DOI: 10.1186/s40164-024-00541-3] [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: 06/11/2024] [Accepted: 07/19/2024] [Indexed: 08/04/2024] Open
Abstract
1q jumping translocation (JT) is rare and its molecular profiles in myeloid malignancies are not well-known. This study evaluated gene mutations in 1q-JT cohorts (0.38%) from hematological malignant specimens that underwent genetic analysis at the Johns Hopkins Hospital (n = 11,908) and the MD Anderson Cancer Center. 1q-JT had frequent mutations in eleven genes, most of which are associated with worse prognosis. BCOR mutations significantly co-occurred with others. Patients tended to have mutations in DNA-repair, spliceosome, and epigenetic modification pathways, though genes utilized within each of these pathways were not randomly distributed. Multi-, albeit overlapping, pathway interruptions tended to manifest in mutations of two gene sets. One gene set consisted of SF3B1 (spliceosome) and TET2 (epigenetic modification), while the other consisted of STAG2 (DNA repair), SRSF2, U2AF (spliceosome), ASXL1, KMT2D (epigenetic modification), BCOR, and GATA2 (transcription factors). An "intermediate" JT-like rearrangement may represent an early sign of occurring 1q-JT. Treatments (hypomethylating agents) and unique structures of the short arms of acrocentric chromosomes may contribute to 1q-JT formation in myeloid malignancies. The median overall survival after identification of a JT was 10 months (95% confidence interval, 5-15 months). Our cohort represents the largest number of myeloid malignancies from multi-centers with before and after the 1q-JT event analyzed to date. Overall, this study identified specific molecular profiles that are associated with 1q-JT in myeloid malignancies. 1q-JT could serve as a poor prognosis biomarker in myeloid malignancies, which could be important in making well-informed clinical decisions and treatment strategies.
Collapse
Affiliation(s)
- Eitan Halper-Stromberg
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Victoria Stinnett
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laura Morsberger
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Aparna Pallavajjala
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mark J Levis
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amy E DeZern
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michelle Lei
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brian Phan
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rena R Xian
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher D Gocke
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Guilin Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, TX, 77030, USA
| | - Ying S Zou
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
6
|
Liu N, Xie Z, Li H, Wang L. The numerous facets of 1q21 + in multiple myeloma: Pathogenesis, clinicopathological features, prognosis and clinical progress (Review). Oncol Lett 2024; 27:258. [PMID: 38646497 PMCID: PMC11027100 DOI: 10.3892/ol.2024.14391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/08/2024] [Indexed: 04/23/2024] Open
Abstract
Multiple myeloma (MM) is a malignant neoplasm characterized by the clonal proliferation of abnormal plasma cells (PCs) in the bone marrow and recurrent cytogenetic abnormalities. The incidence of MM worldwide is on the rise. 1q21+ has been found in ~30-40% of newly diagnosed MM (NDMM) patients.1q21+ is associated with the pathophysiological mechanisms of disease progression and drug resistance in MM. In the present review, the pathogenesis and clinicopathological features of MM patients with 1q21+ were studied, the key data of 1q21+ on the prognosis of MM patients were summarized, and the clinical treatment significance of MM patients with 1q21+ was clarified, in order to provide reference for clinicians to develop treatment strategies targeting 1q21+.
Collapse
Affiliation(s)
- Na Liu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Zhanzhi Xie
- Sanofi China Investment Co., Ltd. Shanghai Branch, Shanghai 200000, P.R. China
| | - Hao Li
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Luqun Wang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
7
|
Caporossi D, Dimauro I. Exercise-induced redox modulation as a mediator of DNA methylation in health maintenance and disease prevention. Free Radic Biol Med 2024; 213:113-122. [PMID: 38242245 DOI: 10.1016/j.freeradbiomed.2024.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 01/21/2024]
Abstract
The evidence for physical activity (PA) as a major public health preventive approach and a potent medical therapy has increased exponentially in the last decades. The biomolecular mechanisms supporting the associations between PA and/or structured exercise training with health maintenance and disease prevention are not completely characterized. However, increasing evidence pointed out the role of epigenetic modifications in exercise adaptation and health-enhancing PA throughout life, DNA methylation being the most intensely studied epigenetic modification induced by acute and chronic exercise. The current data on the modulation of DNA methylation determined by physically active behavior or exercise interventions points out genes related to energy regulation, mitochondrial function, and biosynthesis, as well as muscle regeneration, calcium signaling pathways, and brain plasticity, all consistent with the known exercise-induced redox signaling and/or reactive oxygen species (ROS) unbalance. Thus, the main focus of this review is to discuss the role of ROS and redox-signaling on DNA methylation profile and its impact on exercise-induced health benefits in humans.
Collapse
Affiliation(s)
- Daniela Caporossi
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro De Bosis 15, Rome, 00135, Italy.
| | - Ivan Dimauro
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro De Bosis 15, Rome, 00135, Italy
| |
Collapse
|
8
|
Garcia JB, Storti P, Iannozzi NT, Marchica V, Agnelli L, Toscani D, Franceschi V, Todaro G, Sammarelli G, Notarfranchi L, Scita M, Palma BD, Raimondi V, Lungu O, Pruneri G, Donofrio G, Giuliani N. Identification of PSMB4 and PSMD4 as novel target genes correlated with 1q21 amplification in patients with smoldering myeloma and multiple myeloma. Haematologica 2024; 109:627-631. [PMID: 37608776 PMCID: PMC10828761 DOI: 10.3324/haematol.2023.283200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 08/14/2023] [Indexed: 08/24/2023] Open
Affiliation(s)
| | - Paola Storti
- Department of Medicine and Surgery, University of Parma, Parma
| | | | | | | | - Denise Toscani
- Department of Medicine and Surgery, University of Parma, Parma
| | | | - Giannalisa Todaro
- Hematology and BMT Unit, "Azienda Ospedaliero-Universitaria di Parma", Parma
| | | | | | - Matteo Scita
- Hematology and BMT Unit, "Azienda Ospedaliero-Universitaria di Parma", Parma
| | | | | | - Oxana Lungu
- Department of Medicine and Surgery, University of Parma, Parma
| | | | - Gaetano Donofrio
- Department of Medical-Veterinary Science, University of Parma, Parma
| | - Nicola Giuliani
- Hematology and BMT Unit, "Azienda Ospedaliero-Universitaria di Parma", Parma, Italy; Department of Medicine and Surgery, University of Parma, Parma.
| |
Collapse
|
9
|
Urban VS, Cegledi A, Mikala G. Multiple myeloma, a quintessential malignant disease of aging: a geroscience perspective on pathogenesis and treatment. GeroScience 2022; 45:727-746. [PMID: 36508077 PMCID: PMC9742673 DOI: 10.1007/s11357-022-00698-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 11/18/2022] [Indexed: 12/14/2022] Open
Abstract
Multiple myeloma (MM) is an incurable plasma cell malignancy, which is predominantly a disease of older adults (the median age at diagnosis is 70 years). The slow progression from asymptomatic stages and the late-onset of MM suggest fundamental differences compared to many other hematopoietic system-related malignancies. The concept discussed in this review is that age-related changes at the level of terminally differentiated plasma cells act as the main risk factors for the development of MM. Epigenetic and genetic changes that characterize both MM development and normal aging are highlighted. The relationships between cellular aging processes, genetic mosaicism in plasma cells, and risk for MM and the stochastic processes contributing to clonal selection and expansion of mutated plasma cells are investigated. In line with the DNA damage accumulation theory of aging, in this review, the evolution of monoclonal gammopathy to symptomatic MM is considered. Therapeutic consequences of age-dependent comorbidities that lead to frailty and have fundamental influence on treatment outcome are described. The importance of considering geriatric states when planning the life-long treatment course of an elderly MM patient in order to achieve maximal therapeutic benefit is emphasized.
Collapse
Affiliation(s)
- Veronika S. Urban
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - Andrea Cegledi
- Department of Hematology and Stem Cell Transplantation, South Pest Central Hospital–National Institute for Hematology and Infectious Diseases, Budapest, Hungary
| | - Gabor Mikala
- Department of Hematology and Stem Cell Transplantation, South Pest Central Hospital-National Institute for Hematology and Infectious Diseases, Budapest, Hungary.
| |
Collapse
|
10
|
Chromosomal Heteromorphisms and Cancer Susceptibility Revisited. Cells 2022; 11:cells11203239. [PMID: 36291106 PMCID: PMC9600968 DOI: 10.3390/cells11203239] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/10/2022] [Accepted: 10/10/2022] [Indexed: 11/21/2022] Open
Abstract
Chromosomal heteromorphisms (CHs) are a part of genetic variation in man. The past literature largely posited whether CHs could be correlated with the development of malignancies. While this possibility seemed closed by end of the 1990s, recent data have raised the question again on the potential influences of repetitive DNA elements, the main components of CHs, in cancer susceptibility. Such new evidence for a potential role of CHs in cancer can be found in the following observations: (i) amplification and/or epigenetic alterations of CHs are routinely reported in tumors; (ii) the expression of CH-derived RNA in embryonal and other cells under stress, including cancer cells; (iii) the expression of parts of CH-DNA as long noncoding RNAs; plus (iv) theories that suggest a possible application of the “two-hit model” for euchromatic copy number variants (CNVs). Herein, these points are discussed in detail, which leads to the conclusion that CHs are by far not given sufficient consideration in routine cytogenetic analysis, e.g., leukemias and lymphomas, and need more attention in future research settings including solid tumors. This heightened focus may only be achieved by approaches other than standard sequencing or chromosomal microarrays, as these techniques are at a minimum impaired in their ability to detect, if not blind to, (highly) repetitive DNA sequences.
Collapse
|
11
|
Liu X, Jia S, Chu Y, Tian B, Gao Y, Zhang C, Zheng Y, Jia W, Liu X, Yuan R, Zhang N, Feng J, Dong H, Xin X, Chang Z, Cao Z, Tang H, Gao G. Chromosome 1q21 gain is an adverse prognostic factor for newly diagnosed multiple myeloma patients treated with bortezomib-based regimens. Front Oncol 2022; 12:938550. [PMID: 36185185 PMCID: PMC9516279 DOI: 10.3389/fonc.2022.938550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Chromosome 1q21 aberration is one of the most common cytogenetic abnormalities in multiple myeloma, and is considered an important prognostic factor. The present study analyzed the clinical relevance and prognostic impact of 1q21 gain in 194 patients with newly diagnosed multiple myeloma treated with bortezomib-based regimens. 1q21 gain was detected in 45.9% (89/194) of patients, and those with 1q21 gain had a worse prognosis. Strikingly, our results showed that excluding the effects of other coinciding genetic anomalies, patients carrying at least four copies of 1q21 had worse survival outcome. Moreover, del(13q) strongly correlates with 1q21 gain, and the coexistence of del(13q) and 1q21 gain plays an important role in reducing PFS and OS times. Therefore, 1q21 gain should be considered a high-risk feature in multiple myeloma patients treated with a bortezomib-based regimen.
Collapse
|
12
|
Sklavenitis-Pistofidis R, Getz G, Ghobrial I, Papaioannou M. Multiple Myeloma With Amplification of Chr1q: Therapeutic Opportunity and Challenges. Front Oncol 2022; 12:961421. [PMID: 35912171 PMCID: PMC9331166 DOI: 10.3389/fonc.2022.961421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple myeloma (MM) is an incurable plasma cell malignancy with a heterogeneous genetic background. Each MM subtype may have its own therapeutic vulnerabilities, and tailored therapy could improve outcomes. However, the cumulative frequency of druggable targets across patients is very low, which has precluded the widespread adoption of precision therapy for patients with MM. Amplification of the long arm of chromosome 1 (Amp1q) is one of the most frequent genetic alterations observed in patients with MM, and its presence predicts inferior outcomes in the era of proteasome inhibitors and immunomodulatory agents. Therefore, establishing precision medicine for MM patients with Amp1q stands to benefit a large portion of patients who are otherwise at higher risk of relapse. In this article, we review the prevalence and clinical significance of Amp1q in patients with MM, its pathogenesis and therapeutic vulnerabilities, and discuss the opportunities and challenges for Amp1q-targeted therapy.
Collapse
Affiliation(s)
- Romanos Sklavenitis-Pistofidis
- Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Gad Getz
- Harvard Medical School, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- Department of Pathology, Massachusetts General Hospital, Boston, MA, United States
| | - Irene Ghobrial
- Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Maria Papaioannou
- Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Hematology Unit, 1st Internal Medicine Department, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
13
|
Krhovska P, Pika T, Proskova J, Balcarkova J, Zapletalova J, Bacovsky J, Minarik J. Bone metabolism parameters and their relation to cytogenetics in multiple myeloma. Eur J Haematol Suppl 2022; 109:75-82. [PMID: 35306691 DOI: 10.1111/ejh.13771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/11/2022] [Accepted: 03/16/2022] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Our aim was to correlate serum levels of selected markers of bone metabolism and bone marrow microenvironment to cytogenetic changes in patients with multiple myeloma (MM). METHODS We assed cytogenetic changes in 308 patients and correlated them with the following levels of bone marrow metabolism: thymidine kinase (TK), β2-microglobulin (b-2-m), Dickkopf-1 protein (DKK-1), C-terminal telopeptide collagen-I (ICTP), N-terminal propeptide of type I procollagen (PINP), receptor for interleukin 6 (rIL-6), vascular cell adhesive molecule-1 (VCAM), soluble intercellular adhesion molecule-1, osteoprotegerin (OPG), hepatocyte growth factor (HGF), vascular endothelial growth factor (VEGF), syndecan-1 (SYN-1) and Fas antigen. RESULT Individuals with delRB1 had lower levels of OPG (M = 7.39 vs. 5.46 pmol/L, p = .025) and VEGF (M = 304 vs. 196 pg/ml; p = .036). t(14;16) was associated with higher β2m levels (M = 7.59 vs. 4.13 mg/L; p = .022) and lower DKK-1 levels (M = 4465 ng/L vs. 12,593). The presence of 1q21 gain was associated with higher levels of TK (M = 100.0 vs. 11.0 IU/L, p = .026) and lower levels of PINP (M = 49.3 vs. 67.4 mg/L, p = .030). CONCLUSIONS Our analysis has shown, some cytogenetic changes, especially delRB1, t(14;16) and 1q21gain, which affect the components of the cytokine network in multiple myeloma.
Collapse
Affiliation(s)
- Petra Krhovska
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Tomas Pika
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Jitka Proskova
- Department of Clinical Biochemistry, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Jana Balcarkova
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Jana Zapletalova
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Jaroslav Bacovsky
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Jiri Minarik
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| |
Collapse
|
14
|
Derrien J, Guérin-Charbonnel C, Gaborit V, Campion L, Devic M, Douillard E, Roi N, Avet-Loiseau H, Decaux O, Facon T, Mallm JP, Eils R, Munshi NC, Moreau P, Herrmann C, Magrangeas F, Minvielle S. The DNA methylation landscape of multiple myeloma shows extensive inter- and intrapatient heterogeneity that fuels transcriptomic variability. Genome Med 2021; 13:127. [PMID: 34372935 PMCID: PMC8351364 DOI: 10.1186/s13073-021-00938-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 07/09/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Cancer evolution depends on epigenetic and genetic diversity. Historically, in multiple myeloma (MM), subclonal diversity and tumor evolution have been investigated mostly from a genetic perspective. METHODS Here, we performed an analysis of 42 MM samples from 21 patients by using enhanced reduced representation bisulfite sequencing (eRRBS). We combined several metrics of epigenetic heterogeneity to analyze DNA methylation heterogeneity in MM patients. RESULTS We show that MM is characterized by the continuous accumulation of stochastic methylation at the promoters of development-related genes. High combinatorial entropy change is associated with poor outcomes in our pilot study and depends predominantly on partially methylated domains (PMDs). These PMDs, which represent the major source of inter- and intrapatient DNA methylation heterogeneity in MM, are linked to other key epigenetic aberrations, such as CpG island (CGI)/transcription start site (TSS) hypermethylation and H3K27me3 redistribution as well as 3D organization alterations. In addition, transcriptome analysis revealed that intratumor methylation heterogeneity was associated with low-level expression and high variability. CONCLUSIONS We propose that disrupted DNA methylation in MM is responsible for high epigenetic and transcriptomic instability allowing tumor cells to adapt to environmental changes by tapping into a pool of evolutionary trajectories.
Collapse
Affiliation(s)
- Jennifer Derrien
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, F-44000, France
| | - Catherine Guérin-Charbonnel
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, F-44000, France
- Institut de Cancérologie de l'Ouest, Nantes-Saint Herblain, France
| | - Victor Gaborit
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, F-44000, France
- LS2N, CNRS, Université de Nantes, Nantes, France
| | - Loïc Campion
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, F-44000, France
- Institut de Cancérologie de l'Ouest, Nantes-Saint Herblain, France
| | - Magali Devic
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, F-44000, France
- Centre Hospitalier Universitaire, Nantes, France
| | - Elise Douillard
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, F-44000, France
- Centre Hospitalier Universitaire, Nantes, France
| | - Nathalie Roi
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, F-44000, France
- Centre Hospitalier Universitaire, Nantes, France
| | - Hervé Avet-Loiseau
- Institut Universitaire du Cancer, CHU, Centre de Recherche en Cancérologie de Toulouse, INSERM 1037, Toulouse, France
| | | | | | - Jan-Philipp Mallm
- Research Group Genome Organization & Function, DKFZ, and BioQuant Heidelberg, Heidelberg, 69120, Germany
| | - Roland Eils
- Health Data Science Unit, Medical Faculty Heidelberg and BioQuant, Heidelberg, 69120, Germany
- Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt- Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, Berlin, 10117, Germany
- Berlin Institute of Health (BIH), Center for Digital Health, Anna-Louisa-Karsch-Strasse 2, Berlin, 10178, Germany
| | - Nikhil C Munshi
- Dana-Farber Cancer Institute, Harvard Medical School, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Boston, MA, United States
| | - Philippe Moreau
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, F-44000, France
- Centre Hospitalier Universitaire, Nantes, France
| | - Carl Herrmann
- Health Data Science Unit, Medical Faculty Heidelberg and BioQuant, Heidelberg, 69120, Germany
| | - Florence Magrangeas
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, F-44000, France
- Centre Hospitalier Universitaire, Nantes, France
| | - Stéphane Minvielle
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, F-44000, France.
- Centre Hospitalier Universitaire, Nantes, France.
| |
Collapse
|
15
|
Hanamura I. Gain/Amplification of Chromosome Arm 1q21 in Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13020256. [PMID: 33445467 PMCID: PMC7827173 DOI: 10.3390/cancers13020256] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/07/2021] [Accepted: 01/09/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Multiple myeloma (MM), a plasma cell neoplasm, is an incurable hematological malignancy. Gain/amplification of chromosome arm 1q21 (1q21+) is the most common adverse genomic abnormality associated with disease progression and drug resistance. While possible mechanisms of 1q21+ occurrence and candidate genes in the 1q21 amplicon have been suggested, the precise pathogenesis of MM with 1q21+ is unknown. Herein, we review the current knowledge about the clinicopathological features of 1q21+ MM, which can assist in effective therapeutic approaches for MM patients with 1q21+. Abstract Multiple myeloma (MM), a plasma cell neoplasm, is an incurable hematological malignancy characterized by complex genetic and prognostic heterogeneity. Gain or amplification of chromosome arm 1q21 (1q21+) is the most frequent adverse chromosomal aberration in MM, occurring in 40% of patients at diagnosis. It occurs in a subclone of the tumor as a secondary genomic event and is more amplified as the tumor progresses and a risk factor for the progression from smoldering multiple myeloma to MM. It can be divided into either 1q21 gain (3 copies) or 1q21 amplification (≥4 copies), and it has been suggested that the prognosis is worse in cases of amplification than gain. Trisomy of chromosome 1, jumping whole-arm translocations of chromosome1q, and tandem duplications lead to 1q21+ suggesting that its occurrence is not consistent at the genomic level. Many studies have reported that genes associated with the malignant phenotype of MM are situated on the 1q21 amplicon, including CKS1B, PSMD4, MCL1, ANP32E, and others. In this paper, we review the current knowledge regarding the clinical features, prognostic implications, and the speculated pathology of 1q21+ in MM, which can provide clues for an effective treatment approach to MM patients with 1q21+.
Collapse
Affiliation(s)
- Ichiro Hanamura
- Division of Hematology, Department of Internal Medicine, Aichi Medical University School of Medicine, 1-1, Karimata, Yazako, Nagakute, Aichi 480-1195, Japan
| |
Collapse
|
16
|
Wirth M, Schick M, Keller U, Krönke J. Ubiquitination and Ubiquitin-Like Modifications in Multiple Myeloma: Biology and Therapy. Cancers (Basel) 2020; 12:cancers12123764. [PMID: 33327527 PMCID: PMC7764993 DOI: 10.3390/cancers12123764] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Multiple myeloma is a cancer of plasma cells causing bone fractures, anemia, renal insufficiency and hypercalcemia. Despite the introduction of new drugs in the past years, it still remains incurable and most patients die from the disease. Multiple myeloma cells are characterized by the production of high amounts of monoclonal antibodies. Therefore, maintaining protein homeostasis from synthesis through folding to degradation is crucial for multiple myeloma cells. While protein ubiquitination and organized degradation are typically considered critical for cellular health, an emerging strategy is to block these processes to induce cell death in disease-state cells characterized by protein over-production. Recent development of compounds that alter the ubiquitin proteasome pathway and drugs that affect ubiquitin-like modifications appear promising in both preclinically and in clinical trials. This review summarizes the impact of protein modifications such as ubiquitination and ubiquitin-like modifications in the biology of multiple myeloma and how it can be exploited to develop new effective therapies for multiple myeloma. Abstract Multiple myeloma is a genetically heterogeneous plasma cell malignancy characterized by organ damage and a massive production of (in-)complete monoclonal antibodies. Coping with protein homeostasis and post-translational regulation is therefore essential for multiple myeloma cells to survive. Furthermore, post-translational modifications such as ubiquitination and SUMOylation play key roles in essential pathways in multiple myeloma, including NFκB signaling, epigenetic regulation, as well as DNA damage repair. Drugs modulating the ubiquitin–proteasome system, such as proteasome inhibitors and thalidomide analogs, are approved and highly effective drugs in multiple myeloma. In this review, we focus on ubiquitin and ubiquitin-like modifications in the biology and current developments of new treatments for multiple myeloma.
Collapse
Affiliation(s)
- Matthias Wirth
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 12203 Berlin, Germany; (M.W.); (M.S.); (U.K.)
| | - Markus Schick
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 12203 Berlin, Germany; (M.W.); (M.S.); (U.K.)
| | - Ulrich Keller
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 12203 Berlin, Germany; (M.W.); (M.S.); (U.K.)
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- Max-Delbrück Center for Molecular Medicine, 13092 Berlin, Germany
| | - Jan Krönke
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 12203 Berlin, Germany; (M.W.); (M.S.); (U.K.)
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +49-30-450-513-538
| |
Collapse
|
17
|
Gjerstorff MF. Novel Insights Into Epigenetic Reprogramming and Destabilization of Pericentromeric Heterochromatin in Cancer. Front Oncol 2020; 10:594163. [PMID: 33251148 PMCID: PMC7674669 DOI: 10.3389/fonc.2020.594163] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/15/2020] [Indexed: 12/17/2022] Open
Abstract
Pericentromeric heterochromatin is maintained in a condensed structure by repressive epigenetic control mechanisms and perturbation of these may cause diseases. The chromosome 1q12 region harbors the largest pericentromeric heterochromatin domain in the genome and is among the most common breakpoints in both solid and hematopoietic cancers. Furthermore, the 1q arm is frequently amplified in cancer and this may support tumorigenesis by increasing the dosage of the many oncogenes of this genomic region. Recent studies have provided insight into the mechanisms leading to loss of 1q12 stability and 1q amplification and DNA hypomethylation seems to play a prominent role. This may be the result of decreased activity of DNA methyltransferases and instrumental for 1q12 destabilization or arise secondary to perturbation of other important epigenetic mechanisms that control repression of pericentromeric heterochromatin. Polycomb proteins were recently demonstrated to epigenetically reprogram demethylated 1q12 pericentromeric heterochromatin in premalignant and malignant cells to form large subnuclear structures known as polycomb bodies. This may influence the regulation and stability of 1q12 pericentromeric heterochromatin and/or the distribution of polycomb factors to support tumorigenesis. This review will discuss recent insight into the epigenetic perturbations causing the destabilization of 1q12 pericentromeric heterochromatin and its possible implications for tumor biology.
Collapse
Affiliation(s)
- Morten Frier Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Department of Oncology, Odense University Hospital, Odense, Denmark.,Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, Odense, Denmark
| |
Collapse
|
18
|
Beksac M, Balli S, Akcora Yildiz D. Drug Targeting of Genomic Instability in Multiple Myeloma. Front Genet 2020; 11:228. [PMID: 32373151 PMCID: PMC7179656 DOI: 10.3389/fgene.2020.00228] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 02/26/2020] [Indexed: 12/16/2022] Open
Abstract
Genomic instability can be observed at both chromosomal and chromatin levels. Instability at the macro level includes centrosome abnormalities (CA) resulting in numerical as well as structural chromosomal changes, whereas instability at the micro level is characterized by defects in DNA repair pathways resulting in microsatellite instability (MIN) or mutations. Genomic instability occurs during carcinogenesis without impairing survival and growth, though the precise mechanisms remain unclear. Solid tumors arising from most cells of epithelial origin are characterized by genomic instability which renders them resistant to chemotherapy and radiotherapy. This instability is also observed in 25% of myeloma patients and has been shown to be highly prognostic, independently of the international staging system (ISS). However, a biomarker of aberrant DNA repair and loss of heterozygosity (LOH), was only observed at a frequency of 5% in newly diagnosed patients. Several new molecules targeting the pathways involved in genomic instability are under development and some have already entered clinical trials. Poly(ADP-ribose) polymerase-1 (PARP) inhibitors have been FDA-approved for the treatment of breast cancer type 1 susceptibility protein (BRCA1)-mutated metastatic breast cancer, as well as ovarian and lung cancer. Topoisomerase inhibitors and epigenetic histone modification-targeting inhibitors, such as HDAC (Histone Deacetylase) inhibitors which are novel agents that can target genomic instability. Several of the small molecule inhibitors targeting chromosomal level instability such as PARP, Akt, Aurora kinase, cyclin dependent kinase or spindle kinase inhibitors have been tested in mouse models and early phase I/II trials. ATM, ATR kinase inhibitors and DNA helicase inhibitors are also promising novel agents. However, most of these drugs are not effective as single agents but appear to act synergistically with DNA damaging agents such as radiotherapy, platinum derivatives, immunomodulators, and proteasome inhibitors. In this review, new drugs targeting genomic instability and their mechanisms of action will be discussed.
Collapse
Affiliation(s)
- Meral Beksac
- Department of Hematology, School of Medicine, Ankara University, Ankara, Turkey
| | - Sevinc Balli
- Kars Selim Public Hospital, Internal Medicine, Kars, Turkey
| | - Dilara Akcora Yildiz
- Department of Biology, Science & Art Faculty, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| |
Collapse
|
19
|
Ferreira B, Caetano J, Barahona F, Lopes R, Carneiro E, Costa-Silva B, João C. Liquid biopsies for multiple myeloma in a time of precision medicine. J Mol Med (Berl) 2020; 98:513-525. [PMID: 32246161 PMCID: PMC7198642 DOI: 10.1007/s00109-020-01897-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/06/2020] [Accepted: 03/11/2020] [Indexed: 12/17/2022]
Abstract
Multiple myeloma (MM) is a challenging, progressive, and highly heterogeneous hematological malignancy. MM is characterized by multifocal proliferation of neoplastic plasma cells in the bone marrow (BM) and sometimes in extramedullary organs. Despite the availability of novel drugs and the longer median overall survival, some patients survive more than 10 years while others die rapidly. This heterogeneity is mainly driven by biological characteristics of MM cells, including genetic abnormalities. Disease progressions are mainly due to the inability of drugs to overcome refractory disease and inevitable drug-resistant relapse. In clinical practice, a bone marrow biopsy, mostly performed in one site, is still used to access the genetics of MM. However, BM biopsy use is limited by its invasive nature and by often not accurately reflecting the mutational profile of MM. Recent insights into the genetic landscape of MM provide a valuable opportunity to implement precision medicine approaches aiming to enable better patient profiling and selection of targeted therapies. In this review, we explore the use of the emerging field of liquid biopsies in myeloma patients considering current unmet medical needs, such as assessing the dynamic mutational landscape of myeloma, early predictors of treatment response, and a less invasive response monitoring.
Collapse
Affiliation(s)
- Bruna Ferreira
- Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Joana Caetano
- Hemato-Oncology Unit, Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Filipa Barahona
- Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Raquel Lopes
- Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Emilie Carneiro
- Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Bruno Costa-Silva
- Systems Oncology Group, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Cristina João
- Hemato-Oncology Unit, Myeloma and Lymphoma Research Programme, Nova Medical School, Champalimaud Centre for the Unknown, Lisbon, Portugal.
| |
Collapse
|
20
|
Chatonnet F, Pignarre A, Sérandour AA, Caron G, Avner S, Robert N, Kassambara A, Laurent A, Bizot M, Agirre X, Prosper F, Martin-Subero JI, Moreaux J, Fest T, Salbert G. The hydroxymethylome of multiple myeloma identifies FAM72D as a 1q21 marker linked to proliferation. Haematologica 2020; 105:774-783. [PMID: 31221779 PMCID: PMC7049362 DOI: 10.3324/haematol.2019.222133] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/19/2019] [Indexed: 01/16/2023] Open
Abstract
Cell identity relies on the cross-talk between genetics and epigenetics and their impact on gene expression. Oxidation of 5-methylcytosine (5mC) into 5-hydroxymethylcytosine (5hmC) is the first step of an active DNA demethylation process occurring mainly at enhancers and gene bodies and, as such, participates in processes governing cell identity in normal and pathological conditions. Although genetic alterations are well documented in multiple myeloma (MM), epigenetic alterations associated with this disease have not yet been thoroughly analyzed. To gain insight into the biology of MM, genome-wide 5hmC profiles were obtained and showed that regions enriched in this modified base overlap with MM enhancers and super enhancers and are close to highly expressed genes. Through the definition of a MM-specific 5hmC signature, we identified FAM72D as a poor prognostic gene located on 1q21, a region amplified in high risk myeloma. We further uncovered that FAM72D functions as part of the FOXM1 transcription factor network controlling cell proliferation and survival and we evidenced an increased sensitivity of cells expressing high levels of FOXM1 and FAM72 to epigenetic drugs targeting histone deacetylases and DNA methyltransferases.
Collapse
Affiliation(s)
- Fabrice Chatonnet
- Université Rennes 1, Établissement Français du Sang de Bretaggne, Inserm, MICMAC -UMR_S 1236, Rennes, France
- Laboratoire d'Hématologie, Pôle de Biologie, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Amandine Pignarre
- Université Rennes 1, Établissement Français du Sang de Bretaggne, Inserm, MICMAC -UMR_S 1236, Rennes, France
- Laboratoire d'Hématologie, Pôle de Biologie, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Aurélien A Sérandour
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
- Ecole Centrale de Nantes, Nantes, France
- Institut de Cancérologie de l'Ouest, Site René-Gauducheau, Saint-Herblain, France
| | - Gersende Caron
- Université Rennes 1, Établissement Français du Sang de Bretaggne, Inserm, MICMAC -UMR_S 1236, Rennes, France
- Laboratoire d'Hématologie, Pôle de Biologie, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Stéphane Avner
- SPARTE, IGDR, CNRS UMR6290, University Rennes 1, Rennes, France
| | - Nicolas Robert
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
| | | | - Audrey Laurent
- SPARTE, IGDR, CNRS UMR6290, University Rennes 1, Rennes, France
| | - Maud Bizot
- SPARTE, IGDR, CNRS UMR6290, University Rennes 1, Rennes, France
| | - Xabier Agirre
- Area de Oncología, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Felipe Prosper
- Area de Oncología, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | | | - Jérôme Moreaux
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
- IGH, CNRS, Univ Montpellier, France
| | - Thierry Fest
- Université Rennes 1, Établissement Français du Sang de Bretaggne, Inserm, MICMAC -UMR_S 1236, Rennes, France
- Laboratoire d'Hématologie, Pôle de Biologie, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Gilles Salbert
- SPARTE, IGDR, CNRS UMR6290, University Rennes 1, Rennes, France
| |
Collapse
|
21
|
Kunacheewa C, Lee HC, Patel K, Thomas S, Amini B, Srour S, Bashir Q, Nieto Y, Qazilbash MH, Weber DM, Feng L, Orlowski RZ, Lin P, Manasanch EE. Minimal Residual Disease Negativity Does Not Overcome Poor Prognosis in High-Risk Multiple Myeloma: A Single-Center Retrospective Study. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e221-e238. [PMID: 32037287 DOI: 10.1016/j.clml.2020.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 12/28/2019] [Accepted: 01/02/2020] [Indexed: 01/14/2023]
Abstract
BACKGROUND Minimal residual disease (MRD) is a standard measurement for response assessment in multiple myeloma (MM). Despite new treatments, high-risk MM patients continue to have poor prognosis. We evaluated the effect of MRD negativity in high-risk versus standard-risk patients. PATIENTS AND METHODS We retrospectively evaluated all consecutive MM patients who underwent routine MRD testing by 1-tube 8-color advanced flow cytometry with 2,000,000 events and sensitivity level 10-5 at our center from 2015 to 2018 after initial therapy. Kaplan-Meier and log-rank test were used to assess survival estimates and differences between study groups. RESULTS One hundred thirty-six patients with MRD testing after initial therapy or autologous stem-cell transplantation were identified. At a median follow-up of 14 months (range, 1-36 months), progression-free survival and overall survival were significantly worse in high-risk versus standard-risk patients. During the study period, 50% of high-risk group had experienced disease progression (relapse and/or death) versus 20% in the standard-risk group (P = .0006). No patients with standard-risk died, but 4 (14%) in the high-risk group did (P = .0007). Regardless of MRD status, high-risk patients had statistically significant worse progression-free survival than standard-risk patients. At median follow-up, those with disease 10% standard-risk/MRD negative; 20% standard-risk/MRD positive; 40% high-risk/MRD negative; and 45% high-risk/MRD positive had either experienced relapse or died (P = .0041). MRD status did not significantly affect overall survival in either group (P = .0914); however, longer follow-up is needed to assess survival. CONCLUSION Genetic abnormalities remain a powerful prognostic indicator for MM, regardless of MRD status. For newly diagnosed MM patients treated with novel triple-drug initial therapy and frontline autologous stem-cell transplantation, MRD-negative status did not mitigate the poor-prognosis outcomes of high-risk MM patients.
Collapse
Affiliation(s)
- Chutima Kunacheewa
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hans C Lee
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Krina Patel
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sheeba Thomas
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Behrang Amini
- Division of Diagnostic Imaging, Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Samer Srour
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Qaiser Bashir
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yago Nieto
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Muzzaffar H Qazilbash
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Donna M Weber
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lei Feng
- Department of Statistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Robert Z Orlowski
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Pei Lin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Elisabet E Manasanch
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
22
|
Du C, Mao X, Xu Y, Yan Y, Yuan C, Du X, Liu J, Fan H, Wang Q, Sui W, Deng S, Fu M, Li Z, Li C, Zhao J, Yi S, Liu L, Hao M, Zou D, Zhao Y, Qiu L, An G. 1q21 gain but not t(4;14) indicates inferior outcomes in multiple myeloma treated with bortezomib. Leuk Lymphoma 2019; 61:1201-1210. [PMID: 31842644 DOI: 10.1080/10428194.2019.1700503] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Chenxing Du
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Xuehan Mao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yan Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yuting Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Chenglu Yuan
- Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Xin Du
- Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Jiahui Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Huishou Fan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Qi Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Weiwei Sui
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Shuhui Deng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Mingwei Fu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Zengjun Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Chengwen Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Jiawei Zhao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Shuhua Yi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Lanting Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Mu Hao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Dehui Zou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yaozhong Zhao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Gang An
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Qilu Hospital of Shandong University (Qingdao), Qingdao, China
- Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
23
|
Hu JJ, Nie SM, Gao Y, Yan XS, Huang JX, Li TL, Liu SS, Mao CX, Zhou JJ, Xu YJ, Wang W, Meng FJ, Feng XQ. [The correlations and prognostic value of neutrophil to lymphocyte ratio, immunophenotype and cytogenetic abnormalities in patients with newly diagnosed multiple myeloma]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 40:1044-1046. [PMID: 32023739 PMCID: PMC7342691 DOI: 10.3760/cma.j.issn.0253-2727.2019.12.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Indexed: 11/17/2022]
Affiliation(s)
- J J Hu
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - S M Nie
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Y Gao
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - X S Yan
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - J X Huang
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - T L Li
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - S S Liu
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - C X Mao
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - J J Zhou
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Y J Xu
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - W Wang
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - F J Meng
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - X Q Feng
- Department of Hematology, the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| |
Collapse
|
24
|
Lee I, Gudipati MA, Waters E, Duong VH, Baer MR, Zou Y. Jumping translocations of chromosome 1q occurring by a multi-stage process in an acute myeloid leukemia progressed from myelodysplastic syndrome with a TET2 mutation. Mol Cytogenet 2019; 12:47. [PMID: 31827620 PMCID: PMC6862801 DOI: 10.1186/s13039-019-0460-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 11/01/2019] [Indexed: 11/17/2022] Open
Abstract
Background Jumping translocations (JTs) are rare chromosome rearrangements characterized by re-localization of one donor chromosome to multiple recipient chromosomes. Here, we describe an acute myeloid leukemia (AML) that progressed from myelodysplastic syndrome (MDS) in association with acquisition of 1q JTs. The sequence of molecular and cytogenetic changes in our patient may provide a mechanistic model for the generation of JTs in leukemia. Case presentation A 68-year-old man presented with pancytopenia. Bone marrow aspirate and biopsy showed a hypercellular marrow with multilineage dysplasia, consistent with MDS, with no increase in blasts. Karyotype and MDS fluorescence in situ hybridization (FISH) panel were normal. Repeat bone marrow aspirate and biopsy after 8 cycles of azacitidine, with persistent pancytopenia, showed no changes in morphology, and karyotype was again normal. Myeloid mutation panel showed mutations in RUNX1, SRSF2, ASXL1, and TET2. Three years after diagnosis, he developed AML with myelodysplasia-related changes. Karyotype was abnormal, with unbalanced 1q JTs to the short arms of acrocentric chromosomes 14 and 21, leading to gain of 1q. Conclusions Our patient had MDS with pathogenic mutations of the RUNX1, SRSF2, ASXL1, and TET2 genes and developed 1q JTs at the time of progression from MDS to AML. Our data suggest that the formation of 1q JTs involves multiple stages and may provide a mechanistic model for the generation of JTs in leukemia.
Collapse
Affiliation(s)
- Ina Lee
- 1Department of Pathology, University of Maryland School of Medicine, Baltimore, MD USA
| | - Mary A Gudipati
- 1Department of Pathology, University of Maryland School of Medicine, Baltimore, MD USA
| | - Elizabeth Waters
- 1Department of Pathology, University of Maryland School of Medicine, Baltimore, MD USA
| | - Vu H Duong
- 2Department of Medicine, University of Maryland School of Medicine, Baltimore, MD USA.,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD USA
| | - Maria R Baer
- 2Department of Medicine, University of Maryland School of Medicine, Baltimore, MD USA.,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD USA
| | - Ying Zou
- 1Department of Pathology, University of Maryland School of Medicine, Baltimore, MD USA.,University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD USA.,4Department of Pathology, Johns Hopkins University, 1812 Ashland Ave., Suite 200, Room 221, Baltimore, MD 21205 USA
| |
Collapse
|
25
|
Mohan M, Weinhold N, Schinke C, Thanedrarajan S, Rasche L, Sawyer JR, Tian E, Rhee F, Zangari M. Daratumumab in high‐risk relapsed/refractory multiple myeloma patients: adverse effect of chromosome 1q21 gain/amplification and GEP70 status on outcome. Br J Haematol 2019; 189:67-71. [DOI: 10.1111/bjh.16292] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/13/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Meera Mohan
- Myeloma Center University of Arkansas for Medical Sciences Little Rock AR USA
| | - Niels Weinhold
- Myeloma Center University of Arkansas for Medical Sciences Little Rock AR USA
| | - Carolina Schinke
- Myeloma Center University of Arkansas for Medical Sciences Little Rock AR USA
| | | | - Leo Rasche
- Myeloma Center University of Arkansas for Medical Sciences Little Rock AR USA
| | - Jeffrey R. Sawyer
- Myeloma Center University of Arkansas for Medical Sciences Little Rock AR USA
| | - Erming Tian
- Myeloma Center University of Arkansas for Medical Sciences Little Rock AR USA
| | - Frits Rhee
- Myeloma Center University of Arkansas for Medical Sciences Little Rock AR USA
| | - Maurizio Zangari
- Myeloma Center University of Arkansas for Medical Sciences Little Rock AR USA
| |
Collapse
|
26
|
Sawyer JR, Tian E, Walker BA, Wardell C, Lukacs JL, Sammartino G, Bailey C, Schinke CD, Thanendrarajan S, Davies FE, Morgan GJ, Barlogie B, Zangari M, van Rhee F. An acquired high-risk chromosome instability phenotype in multiple myeloma: Jumping 1q Syndrome. Blood Cancer J 2019; 9:62. [PMID: 31399558 PMCID: PMC6689064 DOI: 10.1038/s41408-019-0226-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/05/2019] [Accepted: 07/11/2019] [Indexed: 02/06/2023] Open
Abstract
Patients with multiple myeloma (MM) accumulate adverse copy number aberrations (CNAs), gains of 1q21, and 17p deletions during disease progression. A subset of these patients develops heightened 1q12 pericentromeric instability and jumping translocations of 1q12 (JT1q12), evidenced by increased copy CNAs of 1q21 and losses in receptor chromosomes (RC). To understand the progression of these aberrations we analyzed metaphase cells of 50 patients with ≥4 CNAs of 1q21 by G-banding, locus specific FISH, and spectral karyotyping. In eight patients with ≥5 CNAs of 1q21 we identified a chromosome instability phenotype similar to that found in ICF syndrome (immunodeficiency, centromeric instability, and facial anomalies). Strikingly, the acquired instability phenotype identified in these patients demonstrates the same transient structural aberrations of 1q12 as those found in ICF syndrome, suggesting similar underlying pathological mechanisms. Four types of clonal aberrations characterize this phenotype including JT1q12s, RC deletions, 1q12-21 breakage-fusion-bridge cycle amplifications, and RC insertions. In addition, recurring transient aberrations include 1q12 decondensation and breakage, triradials, and 1q micronuclei. The acquired self-propagating mobile property of 1q12 satellite DNA drives the continuous regeneration of 1q12 duplication/deletion events. For patients demonstrating this instability phenotype, we propose the term "Jumping 1q Syndrome."
Collapse
Affiliation(s)
- Jeffrey R Sawyer
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA. .,Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| | - Erming Tian
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Brian A Walker
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Christopher Wardell
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Janet L Lukacs
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Gael Sammartino
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Clyde Bailey
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Carolina D Schinke
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - Faith E Davies
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Gareth J Morgan
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Bart Barlogie
- Department of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maurizio Zangari
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Frits van Rhee
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
27
|
Toward personalized treatment in multiple myeloma based on molecular characteristics. Blood 2018; 133:660-675. [PMID: 30587529 DOI: 10.1182/blood-2018-09-825331] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 10/30/2018] [Indexed: 12/11/2022] Open
Abstract
To date, the choice of therapy for an individual multiple myeloma patient has been based on clinical factors such as age and comorbidities. The widespread evolution, validation, and clinical utilization of molecular technologies, such as fluorescence in situ hybridization and next-generation sequencing has enabled the identification of a number of prognostic and predictive biomarkers for progression-free survival, overall survival, and treatment response. In this review, we argue that in order to continue to improve myeloma patient outcomes incorporating such biomarkers into the routine diagnostic workup of patients will allow for the use of personalized, biologically based treatments.
Collapse
|
28
|
Jumping Translocations of 1q in Myelodysplastic Syndrome and Acute Myeloid Leukemia: Report of Three Cases and Review of Literature. Case Rep Genet 2018; 2018:8296478. [PMID: 30271640 PMCID: PMC6151220 DOI: 10.1155/2018/8296478] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 08/15/2018] [Indexed: 01/20/2023] Open
Abstract
Jumping translocations of 1q refer to the break-off of chromosome 1q as a donor fusing to two or more recipient chromosomes. We detected jumping translocations of 1q in three patients with initial diagnosis of myelodysplastic syndrome (MDS) and later progression to acute myeloid leukemia (AML). Review of literature found jumping translocations of 1q in 30 reported cases of MDS and AML. The cytogenetic findings from these 33 cases showed that seven cases had a stemline clone and 26 cases had de novo jumping translocations of 1q in which 5% of cell lineages had additional structural rearrangements. In 75% of cases, the 1q donor jumped to the short arm of recipient acrocentric chromosomes. Approximately 82% of the fusions occurred in the telomeric regions of short and long arms and 18% occurred in the pericentric or interstitial regions of recipient chromosomes. Hypomethylation of the donor 1q pericentromeric region and shortened telomeres in recipient chromosomes were associated with the formation of jumping translocations. Jumping translocations of 1q as an indication of chromosomal instability pose high risk for progression of MDS to AML and a poor prognosis. Further understanding of underlying genomic defects and their clinical significance will improve overall treatment and patient care.
Collapse
|
29
|
Abstract
The enhancer of zeste homolog 2 (EZH2) is the enzymatic subunit of the polycomb repressive complex 2 (PRC2) that exerts important functions during normal development as well as disease. PRC2 through EZH2 tri-methylates histone H3 lysine tail residue 27 (H3K27me3), a modification associated with repression of gene expression programs related to stem cell self-renewal, cell cycle, cell differentiation, and cellular transformation. EZH2 is deregulated and subjected to gain of function or loss of function mutations, and hence functions as an oncogene or tumor suppressor gene in a context-dependent manner. The development of highly selective inhibitors against the histone methyltransferase activity of EZH2 has also contributed to insight into the role of EZH2 and PRC2 in tumorigenesis, and their potential as therapeutic targets in cancer. EZH2 can function as an oncogene in multiple myeloma (MM) by repressing tumor suppressor genes that control apoptosis, cell cycle control and adhesion properties. Taken together these findings have raised the possibility that EZH2 inhibitors could be a useful therapeutic modality in MM alone or in combination with other targeted agents in MM. Therefore, we review the current knowledge on the regulation of EZH2 and its biological impact in MM, the anti-myeloma activity of EZH2 inhibitors and their potential as a targeted therapy in MM.
Collapse
Affiliation(s)
- Mohammad Alzrigat
- Division of Hematology and Oncology, Department of Medicine, University of Florida Health Cancer Center, University of Florida, Gainesville, FL 32610, USA;
| | - Helena Jernberg-Wiklund
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, SE-75185 Uppsala, Sweden;
| | - Jonathan D Licht
- Division of Hematology and Oncology, Department of Medicine, University of Florida Health Cancer Center, University of Florida, Gainesville, FL 32610, USA;
| |
Collapse
|
30
|
Morgan GJ, Rasche L. Maintaining therapeutic progress in multiple myeloma by integrating genetic and biological advances into the clinic. Expert Rev Hematol 2018; 11:513-523. [PMID: 29944024 DOI: 10.1080/17474086.2018.1489718] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Utilizing advances in genetic and immunologic analysis to segment and direct treatment is potentially a way of maintaining therapeutic progress toward cure in multiple myeloma (MM). This approach works well using clinical segments but can be optimized using recent genetic and immunologic technologies, which have opened the possibility of enhancing risk stratification and disease subclassification. Areas covered: This position paper discusses strategies to segment myeloma into subgroups with distinct risk profiles and distinct targetable lesions are presented. Expert commentary: Risk stratified treatment of MM is already a clinical reality that can be enhanced by the developmental of unified segmentation and testing approaches. Mutation-targeted treatment has proven to be effective against the RAS pathway, but is compromised by intra-clonal and spatiotemporal heterogeneity. Identifying new disease segments based on tumor biology or immunological content of the microenvironment offers an exciting new way to control and even eradicate myeloma clones. Going forward, risk and biologically stratified therapy for myeloma is a promising way of maintaining therapeutic progress, as is precision immunotherapy directed by the cellular context of the bone marrow.
Collapse
Affiliation(s)
- Gareth J Morgan
- a Myeloma Institute , The University of Arkansas for Medical Sciences , Little Rock , AR , USA
| | - Leo Rasche
- a Myeloma Institute , The University of Arkansas for Medical Sciences , Little Rock , AR , USA
| |
Collapse
|
31
|
Kumar SK, Rajkumar SV. The multiple myelomas — current concepts in cytogenetic classification and therapy. Nat Rev Clin Oncol 2018; 15:409-421. [DOI: 10.1038/s41571-018-0018-y] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
32
|
SETDB1-mediated FosB regulation via ERK2 is associated with an increase in cell invasiveness during anticancer drug treatment of A549 human lung cancer cells. Biochem Biophys Res Commun 2017; 495:512-518. [PMID: 29108991 DOI: 10.1016/j.bbrc.2017.10.176] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 10/29/2017] [Indexed: 02/08/2023]
Abstract
We have determined a functional link to the inverse expression of SETDB1 and FosB following anticancer drug treatment. Doxorubicin treatment caused decreased SETDB1 expression and FosB overexpression both at the mRNA and protein levels. The decreased HMTase activity of SETDB1 coincided with altered occupancy across the promoter region of the FosB gene. SETDB1 overexpression decreased the luciferase reporter activity containing the FosB promoter region, but siSETDB1 increased the luciferase reporter activity, suggesting that SETDB1 directly and negatively regulated FosB expression. In addition, MEK inhibitor (PD98059) blocked the SETDB1 regulation of the FosB promoter activity via ERK2 activation during doxorubicin treatment. A microscopic analysis reveals that FosB expression was observed in living cells in spite of doxorubicin treatment. Ectopic FosB/ΔFosB expression increased the number of colonies and the migration of A549 cells compared to that in control. These results suggest that the ERK2-SETDB1-FosB signaling pathway might have an anti-therapeutic regulatory mechanism that increases the transformation and migration activity of cancer cells during anticancer drug treatment.
Collapse
|
33
|
Ixazomib significantly prolongs progression-free survival in high-risk relapsed/refractory myeloma patients. Blood 2017; 130:2610-2618. [PMID: 29054911 DOI: 10.1182/blood-2017-06-791228] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 10/09/2017] [Indexed: 12/12/2022] Open
Abstract
Certain cytogenetic abnormalities are known to adversely impact outcomes in patients with multiple myeloma (MM). The phase 3 TOURMALINE-MM1 study demonstrated a significant improvement in progression-free survival (PFS) with ixazomib-lenalidomide-dexamethasone (IRd) compared with placebo-lenalidomide-dexamethasone (placebo-Rd). This preplanned analysis assessed the efficacy and safety of IRd vs placebo-Rd according to cytogenetic risk, as assessed using fluorescence in situ hybridization. High-risk cytogenetic abnormalities were defined as del(17p), t(4;14), and/or t(14;16); additionally, patients were assessed for 1q21 amplification. Of 722 randomized patients, 552 had cytogenetic results; 137 (25%) had high-risk cytogenetic abnormalities and 172 (32%) had 1q21 amplification alone. PFS was improved with IRd vs placebo-Rd in both high-risk and standard-risk cytogenetics subgroups: in high-risk patients, the hazard ratio (HR) was 0.543 (95% confidence interval [CI], 0.321-0.918; P = .021), with median PFS of 21.4 vs 9.7 months; in standard-risk patients, HR was 0.640 (95% CI, 0.462-0.888; P = .007), with median PFS of 20.6 vs 15.6 months. This PFS benefit was consistent across subgroups with individual high-risk cytogenetic abnormalities, including patients with del(17p) (HR, 0.596; 95% CI, 0.286-1.243). PFS was also longer with IRd vs placebo-Rd in patients with 1q21 amplification (HR, 0.781; 95% CI, 0.492-1.240), and in the "expanded high-risk" group, defined as those with high-risk cytogenetic abnormalities and/or 1q21 amplification (HR, 0.664; 95% CI, 0.474-0.928). IRd demonstrated substantial benefit compared with placebo-Rd in relapsed and/or refractory MM (RRMM) patients with high-risk and standard-risk cytogenetics, and improves the poor PFS associated with high-risk cytogenetic abnormalities. This trial was registered at www.clinicaltrials.gov as #NCT01564537.
Collapse
|
34
|
Epigenetics in multiple myeloma: From mechanisms to therapy. Semin Cancer Biol 2017; 51:101-115. [PMID: 28962927 DOI: 10.1016/j.semcancer.2017.09.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 08/25/2017] [Accepted: 09/25/2017] [Indexed: 12/22/2022]
Abstract
Multiple myeloma (MM) is a tumor of antibody producing plasmablasts/plasma cells that resides within the bone marrow (BM). In addition to the well-established role of genetic lesions and tumor-microenvironment interactions in the development of MM, deregulated epigenetic mechanisms are emerging as important in MM pathogenesis. Recently, MM sequencing and expression projects have revealed that mutations and copy number variations as well as deregulation in the expression of epigenetic modifiers are characteristic features of MM. In the past decade, several studies have suggested epigenetic mechanisms via DNA methylation, histone modifications and non-coding RNAs as important contributing factors in MM with impacts on disease initiation, progression, clonal heterogeneity and response to treatment. Herein we review the present view and knowledge that has accumulated over the past decades on the role of epigenetics in MM, with focus on the interplay between epigenetic mechanisms and the potential use of epigenetic inhibitors as future treatment modalities for MM.
Collapse
|
35
|
Greil C, Ihorst G, Gaiser F, Salzer U, Bisse E, Kastritis E, Ludwig H, Wäsch R, Engelhardt M. The serum heavy/light chain immunoassay: A valuable tool for sensitive paraprotein assessment, risk, and disease monitoring in monoclonal gammopathies. Eur J Haematol 2017; 99:449-458. [PMID: 28886228 DOI: 10.1111/ejh.12958] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVE The heavy/light chain (HLC)-immunoassay quantifies light chain types of each immunoglobulin class in patients with monoclonal gammopathies. METHODS We assessed 147 consecutive patients with different forms and stages of plasma cell dyscrasias (PCD) who received standard tests (serum and urine protein electrophoresis [SPEP, UPEP], immunofixation [IFE], serum-free light chain [SFLC]), and HLC-immunoassay. Patients with multiple myeloma (MM, n = 102), smoldering MM (SMM, n = 5), monoclonal gammopathy of undetermined significance (MGUS, n = 28), and Waldenström's macroglobulinemia (WM, n = 12) were included. RESULTS We verified a significant correlation between HLC- and standard monoclonal protein (mp)-parameters, and HLC-increases with higher disease stage and unfavorable remission status. In patients with difficult to quantify mp, more abnormal HLC- than SPEP-, immunoglobulin-, or SFLC-results were found. In WM, a pathological HLC κ/λ-ratio and M-component were observed in 95% and 58%, respectively. In 21/28 MGUS and 5/5 SMM patients, HLC κ/λ-ratios were abnormal. Testing different HLC cutoffs, patients with extreme HLC values showed impaired progression-free survival (PFS). CONCLUSIONS Despite the fact that different PCD patients were included, the assessment of the HLC-immunoassay in MGUS, SMM, MM, and WM, our comparison with standard mp-assays, and relevant PFS differences may excite future applications, which should be confirmed in prospective multicenter trials.
Collapse
Affiliation(s)
- Christine Greil
- Department of Medicine I, Hematology, Oncology & Stem Cell Transplantation, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | - Gabriele Ihorst
- Faculty of Medicine, Clinical Trials Unit, University of Freiburg, Freiburg, Germany
| | - Felix Gaiser
- Department of Medicine I, Hematology, Oncology & Stem Cell Transplantation, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | - Ulrich Salzer
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | - Emanuel Bisse
- Central Laboratory, Medical Center, University of Freiburg, Freiburg, Germany
| | | | - Heinz Ludwig
- Zentrum für Onkologie, Wihelminenkrebsforschungsinstitut, Beethovengasse, Österreich
| | - Ralph Wäsch
- Department of Medicine I, Hematology, Oncology & Stem Cell Transplantation, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | - Monika Engelhardt
- Department of Medicine I, Hematology, Oncology & Stem Cell Transplantation, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| |
Collapse
|
36
|
Berry NK, Dixon-McIver A, Scott RJ, Rowlings P, Enjeti AK. Detection of complex genomic signatures associated with risk in plasma cell disorders. Cancer Genet 2017; 218-219:1-9. [PMID: 29153091 DOI: 10.1016/j.cancergen.2017.08.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 03/30/2017] [Accepted: 08/06/2017] [Indexed: 01/10/2023]
Abstract
Plasma cell disorders (PCD) range from benign to highly malignant disease. The ability to detect risk-stratifying aberrations based on cytogenetic and molecular genetic assays plays an increasing role in therapeutic decision making. In this study, 58 patients were chosen for screening by comparative genomic hybridisation microarray (aCGH) to identify the new high-risk prognostic markers of chromothripsis and chromoanasynthesis. All patients had an unequivocal clinical diagnosis of a plasma cell disorder (plasma cell myeloma (PCM)(n = 51) or monoclonal gammopathy of undetermined significance (MGUS)(n = 7)) and an abnormal FISH result. There were a total of 17 complex genomic events identified across 9 patient samples, which were selected for further investigation by high definition single nucleotide polymorphism (HD-SNP) microarray. Each event was analysed and characterised for chromothripsis, chromoanasynthesis or a complex step-wise chromosomal event. We describe an effective method to identify the new high-risk prognostic markers of chromothripsis and chromoanasynthesis in plasma cell disorders.
Collapse
Affiliation(s)
- Nadine K Berry
- Department of Hematology, Calvary Mater Hospital, Newcastle, New South Wales, Australia; School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia; School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia.
| | | | - Rodney J Scott
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia; Department of Molecular Medicine, Pathology North Newcastle, Rankin Park, New South Wales, Australia
| | - Philip Rowlings
- Department of Hematology, Calvary Mater Hospital, Newcastle, New South Wales, Australia; School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia
| | - Anoop K Enjeti
- Department of Hematology, Calvary Mater Hospital, Newcastle, New South Wales, Australia; School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
37
|
Abstract
The outcomes for the majority of patients with myeloma have improved over recent decades, driven by treatment advances. However, there is a subset of patients considered to have high-risk disease who have not benefited. Understanding how high-risk disease evolves from more therapeutically tractable stages is crucial if we are to improve outcomes. This can be accomplished by identifying the genetic mechanisms and mutations driving the transition of a normal plasma cell to one with the features of the following disease stages: monoclonal gammopathy of undetermined significance, smouldering myeloma, myeloma and plasma cell leukaemia. Although myeloma initiating events are clonal, subsequent driver lesions often occur in a subclone of cells, facilitating progression by Darwinian selection processes. Understanding the co-evolution of the clones within their microenvironment will be crucial for therapeutically manipulating the process. The end stage of progression is the generation of a state associated with treatment resistance, increased proliferation, evasion of apoptosis and an ability to grow independently of the bone marrow microenvironment. In this Review, we discuss these end-stage high-risk disease states and how new information is improving our understanding of their evolutionary trajectories, how they may be diagnosed and the biological behaviour that must be addressed if they are to be treated effectively.
Collapse
Affiliation(s)
- Charlotte Pawlyn
- The Institute of Cancer Research, 15 Cotswold Road, Sutton SM2 5NG, UK
| | - Gareth J Morgan
- The Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA
| |
Collapse
|
38
|
Smol T, Dufour A, Tricot S, Wemeau M, Stalnikiewicz L, Bernardi F, Terré C, Ducourneau B, Bisiau H, Daudignon A. Combination of t(4;14), del(17p13), del(1p32) and 1q21 gain FISH probes identifies clonal heterogeneity and enhances the detection of adverse cytogenetic profiles in 233 newly diagnosed multiple myeloma. Mol Cytogenet 2017; 10:26. [PMID: 28680482 PMCID: PMC5493886 DOI: 10.1186/s13039-017-0327-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 06/25/2017] [Indexed: 01/26/2023] Open
Abstract
Background Our aim was to set the FISH combination of del(17p13), t(4;14), 1q21 gain and del(1p32), four adverse cytogenetic factors rarely evaluated together, and compare our technical thresholds with those defined in the literature. Methods Two hundred thirty-three patients with MM at diagnosis were studied using FISH to target 4 unfavorable cytogenetic abnormalities: 17p13 deletion, t(4;14) translocation, 1p32 deletion and 1q21 gain. Technical thresholds were determined for each probe using isolated CD138-expressing PC from patients without MM. Results The FISH analysis identified abnormalities in 79.0% of patients. Del(17p13) was detected in 15.0% of cases, t(4;14) in 11.5%, 1q21 gain in 37.8% and del(1p32) in 8.7%. Adding 1p32/1q21 FISH probes has enabled us to identify adverse cytogenetic profiles in 39.0% of patients without del(17p13) or t(4;14). Clonal heterogeneity was observed in 51.1% of patients as well as an increase in the number of adverse abnormalities when related clones were greater than or equal to 2 (85.1% against 45.6%). Conclusion FISH allowed detecting accumulation of adverse abnormalities and clonal heterogeneity in MM with a combination of 4 probes. The impacts of these two parameters need to be evaluated, and could be included in future cytogenetic classifications.
Collapse
Affiliation(s)
- Thomas Smol
- Service d'Hématologie-Immunologie-Cytogénétique, CH Valenciennes, Valenciennes, France.,Université de Lille Nord de France, Lille, France.,Institut de génétique médicale, Hôpital Jeanne de Flandre, CHRU Lille, Lille, France
| | - Annika Dufour
- Laboratoire de Cytogénétique Hématologique, CH Versailles, Le Chesnay, France
| | - Sabine Tricot
- Service d'Hématologie Clinique, CH Valenciennes, Valenciennes, France
| | | | | | | | - Christine Terré
- Laboratoire de Cytogénétique Hématologique, CH Versailles, Le Chesnay, France
| | - Benoît Ducourneau
- Service d'Hématologie-Immunologie-Cytogénétique, CH Valenciennes, Valenciennes, France
| | - Hervé Bisiau
- Service d'Hématologie-Immunologie-Cytogénétique, CH Valenciennes, Valenciennes, France
| | - Agnès Daudignon
- Service d'Hématologie-Immunologie-Cytogénétique, CH Valenciennes, Valenciennes, France
| |
Collapse
|
39
|
Alzrigat M, Jernberg-Wiklund H. The miR-125a and miR-320c are potential tumor suppressor microRNAs epigenetically silenced by the polycomb repressive complex 2 in multiple myeloma. RNA & DISEASE 2017; 4. [PMID: 28664185 PMCID: PMC5485917 DOI: 10.14800/rd.1529] [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] [Indexed: 12/12/2022] Open
Abstract
We have previously presented the histone methyltransferase enhancer of zeste homolog 2 (EZH2) of the polycomb repressive complex 2 (PRC2) as a potential therapeutic target in Multiple Myeloma (MM). In a recent article in Oncotarget by Alzrigat. et al. 2017, we have reported on the novel finding that EZH2 inhibition using the highly selective inhibitor of EZH2 enzymatic activity, UNC1999, reactivated the expression of microRNA genes previously reported to be underexpressed in MM. Among these, we have identified miR-125a-3p and miR-320c as potential tumor suppressor microRNAs as they were predicted to target MM-associated oncogenes; IRF-4, XBP-1 and BLIMP-1. We also found EZH2 inhibition to reactivate the expression of miR-494, a previously reported regulator of the c-MYC oncogene. In addition, we could report that EZH2 inhibition downregulated the expression of a few well described oncogenic microRNAs in MM. The data from our recent article are here highlighted as it shed a new light onto the oncogenic function of the PRC2 in MM. These data further strengthen the notion that the PRC2 complex may be of potential therapeutic interest.
Collapse
Affiliation(s)
- Mohammad Alzrigat
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, SE-751 85, Sweden
| | - Helena Jernberg-Wiklund
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, SE-751 85, Sweden
| |
Collapse
|
40
|
Morgan GJ, Jones JR. Integration of Genomics Into Treatment: Are We There Yet? Am Soc Clin Oncol Educ Book 2017; 37:569-574. [PMID: 28561666 DOI: 10.1200/edbk_175166] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Using advances in genetic analysis to segment and direct treatment of multiple myeloma (MM) represents a way of maintaining therapeutic progress. Recent genetic analyses have opened the possibility of enhancing risk stratification approaches and of using different risk and biologic strata as part of clinical trials. The Myeloma Genome Project is a collaborative project that has compiled the largest set of cases with sequencing and have outcome data that are available for stratification purposes. Mutation-targeted treatment of the Ras pathway has been shown to be active in MM, but is compromised by the presence of the subclonal genetic variation typical of myeloma. Going forward, risk and biologically stratified therapy for MM looks to be a promising way of maintaining therapeutic progress, as does precision immunotherapy directed by the cellular context of the bone marrow.
Collapse
Affiliation(s)
- Gareth J Morgan
- From the Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR; Institute of Cancer Research, The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
| | - John R Jones
- From the Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR; Institute of Cancer Research, The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
41
|
Gu C, Feng L, Peng H, Yang H, Feng Z, Yang Y. MTDH is an oncogene in multiple myeloma, which is suppressed by Bortezomib treatment. Oncotarget 2016; 7:4559-69. [PMID: 26683226 PMCID: PMC4826226 DOI: 10.18632/oncotarget.6610] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 11/26/2015] [Indexed: 12/19/2022] Open
Abstract
Metadherin (MTDH) is identified as an oncogene in multiple cancers including breast cancer, bladder cancer and endometrial cancer. However, the function of MTDH in multiple myeloma (MM) is still unexplored. In this study, we disclose that MTDH is an oncogene in MM. This is characterized by an elevation mRNA level of MTDH and chromosomal gain of MTDH locus in MM cells compared to normal samples. Moreover, MTDH expression significantly increased in MMSET translocation (MS) subgroup, one of the high-risk subgroups in MM, and was significantly correlated with MM patients' poor outcomes in Total Therapy 2 (TT2) cohort. Further knockdown of MTDH expression by shRNA in MM cells induced cell apoptosis, inhibited MM cells growth in vitro and decreased xenograft tumor formation in vivo. Interestingly, opposite to TT2, MM patients with high-MTDH expression showed favorable survival outcomes in the TT3 cohort, while Bortezomib treatment was the major difference between TT2 and TT3 cohort. Furthermore we proved that Bortezomib suppressed pre- and post-transcription levels of MTDH expression of MM cells in vitro and in vivo. Finally, our studies demonstrated that MTDH is a transcriptional gene of MMSET/NFκB /MYC signaling in MM cells, which is inhibited by Bortezomib treatment.
Collapse
Affiliation(s)
- Chunyan Gu
- Department of Pathology, Nanjing Medical University, 210029, Nanjing, China.,Basic Medical College, Nanjing University of Chinese Medicine, 210046, Nanjing, China.,Department of Pathology, University of Iowa Carver College of Medicine, 52242, Iowa City, IA, USA
| | - Lang Feng
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, 100050, Beijing, China.,Department of Urology, University of Iowa Carver College of Medicine, 52242, Iowa City, IA, USA
| | - Hailin Peng
- Department of Laboratory Medicine, Taizhou people's hospital, 225300, Taizhou, China
| | - Hongbao Yang
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, 211198, China
| | - Zhenqing Feng
- Department of Pathology, Nanjing Medical University, 210029, Nanjing, China
| | - Ye Yang
- Basic Medical College, Nanjing University of Chinese Medicine, 210046, Nanjing, China.,Department of Internal Medicine, University of Iowa Carver College of Medicine, 52242, Iowa City, IA, USA
| |
Collapse
|
42
|
Hyperhaploidy is a novel high-risk cytogenetic subgroup in multiple myeloma. Leukemia 2016; 31:637-644. [PMID: 27694925 DOI: 10.1038/leu.2016.253] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 07/20/2016] [Accepted: 08/19/2016] [Indexed: 12/13/2022]
Abstract
Hyperhaploid clones (24-34 chromosomes) were identified in 33 patients with multiple myeloma (MM), demonstrating a novel numerical cytogenetic subgroup. Strikingly, all hyperhaploid karyotypes were found to harbor monosomy 17p, the single most important risk stratification lesion in MM. A catastrophic loss of nearly a haploid set of chromosomes results in disomies of chromosomes 3, 5, 7, 9, 11, 15, 18, 19 and 21, the same basic set of odd-numbered chromosomes found in trisomy in hyperdiploid myeloma. All other autosomes are found in monosomy, resulting in additional clinically relevant monosomies of 1p, 6q, 13q and 16q. Hypotriploid subclones (58-68 chromosomes) were also identified in 11 of the 33 patients and represent a duplication of the hyperhaploid clone. Analysis of clones utilizing interphase fluorescence in situ hybridization (iFISH), metaphase FISH and spectral karyotyping identified either monosomy 17 or del17p in all patients. Amplification of 1q21 was identified in eight patients, demonstrating an additional high-risk marker. Importantly, our findings indicate that current iFISH strategies may be uninformative or ambiguous in the detection of these clones, suggesting this patient subgroup maybe underreported. Overall survival for patients with hyperhaploid clones was poor, with a 5-year survival rate of 23.1%. These findings identify a distinct numerical subgroup with cytogenetically defined high-risk disease.
Collapse
|
43
|
Rajan AM, Rajkumar SV. Interpretation of cytogenetic results in multiple myeloma for clinical practice. Blood Cancer J 2015; 5:e365. [PMID: 26517360 PMCID: PMC4635200 DOI: 10.1038/bcj.2015.92] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 09/18/2015] [Indexed: 12/16/2022] Open
Abstract
The interpretation of cytogenetic abnormalities in multiple myeloma (MM) is often a challenging task. MM is characterized by several cytogenetic abnormalities that occur at various time points in the disease course. The interpretation of cytogenetic results in MM is complicated by the number and complexity of the abnormalities, the methods used to detect them and the disease stage at which they are detected. Specific cytogenetic abnormalities affect clinical presentation, progression of smoldering multiple myeloma (SMM) to MM, prognosis of MM and management strategies. The goal of this paper is to provide a review of how MM is classified into specific subtypes based on primary cytogenetic abnormalities and to provide a concise overview of how to interpret cytogenetic abnormalities based on the disease stage to aid clinical practice and patient management.
Collapse
Affiliation(s)
- A M Rajan
- Aureus University School of Medicine, Oranjestad, Aruba
| | - S V Rajkumar
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|