1
|
Jha K, Saha S, Bhattacharyya M. Cytogenetic Alterations and Correlation with Age and Gender in Patients of Multiple Myeloma: A Study from a Tertiary Care Center in Eastern India. South Asian J Cancer 2024; 13:126-131. [PMID: 38919660 PMCID: PMC11196154 DOI: 10.1055/s-0043-1761441] [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] [Indexed: 06/27/2024] Open
Abstract
Karuna JhaBackground Multiple myeloma is a cytogenetically heterogeneous, evolving, and incurable disease. Differences in prevalence of myeloma already exist in Indian subcontinent as compared with Western world countries. This study attempts to investigate differences in incidence of cytogenetic abnormalities (CA) in Eastern Indian patients and study differences in incidence with respect to age and gender. Materials and Methods Interphase fluorescence in situ hybridization (FISH) was applied on purified plasma cells of 280 newly diagnosed myeloma cases using specific probes. Statistical Analysis Data was analyzed using SPSS software version 25. Results Note that 51.07% patients were FISH positive. Del13q was the most common CA. Significant association of del 13q with t(4;14), del 17p, and gain of 1q was seen. The frequencies of FISH positive and negative groups differed in the different age groups; higher number of cases in 41 to 50 years group in FISH positive group ( p < 0.05) and lower number of cases in FISH positive group in 61 to 70 years ( p < 0.05) as compared with FISH negative group. Del 17p had higher number of cases in age group 41 to 50 years and 51 to 60 years as compared with other age groups. Incidence of t(11;14) was in 5th to 7th decade while del 13q and t(4;14) had the widest range of age at presentation. Gender disparities were seen in high-risk cytogenetics like del 17p and 1q gain. Conclusion The differences in incidence rate of CAs per se in myeloma cases diagnosed in Indian subcontinent and the differences in incidence with respect to age and gender warrant further multicentric studies.
Collapse
Affiliation(s)
- Karuna Jha
- Department of Clinical Hematology, Institute of Hematology and Transfusion Medicine, Medical College, Kolkata, West Bengal, India
| | - Sandeep Saha
- Department of Clinical Hematology, Institute of Hematology and Transfusion Medicine, Medical College, Kolkata, West Bengal, India
| | - Maitreyee Bhattacharyya
- Department of Clinical Hematology, Institute of Hematology and Transfusion Medicine, Medical College, Kolkata, West Bengal, India
| |
Collapse
|
2
|
Pasvolsky O, Ghanem S, Milton DR, Rauf M, Tanner MR, Bashir Q, Srour S, Saini N, Lin P, Ramdial J, Nieto Y, Tang G, Aljawai Y, Khan HN, Kebriaei P, Lee HC, Patel KK, Thomas SK, Weber DM, Orlowski RZ, Shpall EJ, Champlin RE, Qazilbash MH. Outcomes of patients with multiple myeloma and 1q gain/amplification receiving autologous hematopoietic stem cell transplant: the MD Anderson cancer center experience. Blood Cancer J 2024; 14:4. [PMID: 38199987 PMCID: PMC10781953 DOI: 10.1038/s41408-023-00973-w] [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: 10/02/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
The prognostic impact of additional copies of chromosome 1q (1q + ) on outcomes of newly-diagnosed multiple myeloma (NDMM) patients undergoing autologous transplantation (autoSCT) is unclear. We conducted a retrospective single-center analysis of NDMM patients with 1q21 gain/amplification (3 or ≥4 copies of 1q, respectively) that received autoSCT between 2008-2018. 213 patients were included (79% 1q gain; 21% 1q amplification). The most commonly used induction regimen was bortezomib, lenalidomide, and dexamethasone (41%). At day100 post-autoSCT and at best post-transplant response, 78% and 87% of patients achieved ≥VGPR, and 38% and 50% achieved MRD-negative ≥VGPR, respectively. Median PFS and OS for the entire cohort were 35.5 months and 81.4 months, respectively. On multivariable assessment for PFS, MRD negative ≥VGPR before autoSCT (HR 0.52, p = 0.013) was associated with superior PFS, whereas 1q amplification was associated with inferior PFS (2.03, p = 0.003). On multivariate analysis for OS, achieving MRD negative ≥VGPR at best post-transplant response was associated with superior survival (0.29, p < 0.001), whereas R-ISS III and concomitant del17p or t(4:14) were associated with inferior survival (6.95, p = 0.030, 2.33, p = 0.023 and 3.00, p = 0.047, respectively). In conclusion, patients with 1q+ NDMM, especially 1q amplification, have inferior survival outcomes compared to standard-risk disease after upfront autoSCT, though outcomes are better than other high-risk cytogenetic abnormalities.
Collapse
Affiliation(s)
- Oren Pasvolsky
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sassine Ghanem
- Lifespan Cancer Institute, Providence, RI, USA
- Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Denái R Milton
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mikael Rauf
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark R Tanner
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qaiser Bashir
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Samer Srour
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Neeraj Saini
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul Lin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeremy Ramdial
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yago Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Guilin Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yosra Aljawai
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hina N Khan
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Hematology/Oncology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hans C Lee
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Krina K Patel
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sheeba K Thomas
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Donna M Weber
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert Z Orlowski
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Muzaffar H Qazilbash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
3
|
Kalal AA, Arumugam M, Shetty VV, Shetty KP, Krishna R, Shetty RA, Kulkarni NV, Shetty DP. A diagnostic approach to detect cytogenetic heterogeneity and its prognostic significance in multiple myeloma. J Taibah Univ Med Sci 2023; 18:1138-1147. [PMID: 37206191 PMCID: PMC10189271 DOI: 10.1016/j.jtumed.2023.03.009] [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: 10/02/2022] [Revised: 02/17/2023] [Accepted: 03/16/2023] [Indexed: 03/29/2023] Open
Abstract
Objective Multiple myeloma (MM) is a hematological disorder involving the uncontrolled proliferation of clonal plasma cells and its accumulation in the bone marrow. This study analyzed the frequency, cytogenetic heterogeneity, and clinical characteristics of patients with MM. Methods Bone marrow aspirates were obtained from 72 patients with MM and evaluated by conventional cytogenetics (CCs) and interphase fluorescence in situ hybridization (iFISH) techniques for a panel of probes, including immunoglobulin heavy chain (IgH)/CCND1, IgH/fibroblast growth factor receptor 3 (FGFR3), IgH/MAFB, 13q deletion, and deletion 17p. Results CCs revealed abnormal karyotypes in 39% of the patients examined. The incidence of hypodiploidy was 28% (20/72) while that of hyperdiploidy was 10% (7/72). iFISH analysis revealed t(11;14) in 6% (4/72) and t(4;14) in 11% (8/72) of patients. Patients with hyperdiploidy and hypodiploidy were associated with several monosomies and trisomies. Kaplan-Meier analysis revealed a significant difference between positive and negative groups for t(4;14), trisomy 14, and monosomy 13; this was associated with a shorter survival time. Cox proportional analysis identified t(4;14) (P = 0.032), trisomy 14 (P = 0.004), and monosomy 13 (P = 0.009), as significant factors with hazard ratio of 0.187 [confidence interval (CI): 0.041-0.862], 0.109 [CI: 0.024-0.500] and 0.134 [CI: 0.030-0.600]. Conclusion In addition to cytogenetic abnormalities, iFISH analysis revealed significant heterogeneity among patients with MM. Cytogenetic heterogeneity in patients with MM should be considered as a major prognostic marker contributing to the variability of the disease. Our findings suggest that these abnormalities are independent prognostic factors.
Collapse
Affiliation(s)
- Akanksha A. Kalal
- KSHEMA Center for Genetic Services, KS Hegde Medical Academy, NITTE (Deemed to be University), Mangaluru, Karnataka, India
| | | | - Vijith V. Shetty
- Department of Oncology, KS Hegde Medical Academy, NITTE (Deemed to be University), Mangaluru, Karnataka, India
| | - K. Padma Shetty
- Department of Pathology, KS Hegde Medical Academy, NITTE (Deemed to be University), Mangaluru, Karnataka, India
| | - Rajesh Krishna
- Department of Oncology, Yenepoya Medical College, Yenepoya (Deemed to be University), Mangaluru, Karnataka, India
| | - Reshma A. Shetty
- KSHEMA Center for Genetic Services, KS Hegde Medical Academy, NITTE (Deemed to be University), Mangaluru, Karnataka, India
| | - Nagaraj V. Kulkarni
- KSHEMA Center for Genetic Services, KS Hegde Medical Academy, NITTE (Deemed to be University), Mangaluru, Karnataka, India
| | - D. Prashanth Shetty
- KSHEMA Center for Genetic Services, KS Hegde Medical Academy, NITTE (Deemed to be University), Mangaluru, Karnataka, India
| |
Collapse
|
4
|
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
|
5
|
Thi Vinh Do A, Lan Anh L. CD38 – Negative Anaplastic Plasma Cell Myeloma: A Rare Case Report. Cureus 2022; 14:e20909. [PMID: 35004078 PMCID: PMC8723977 DOI: 10.7759/cureus.20909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2022] [Indexed: 11/19/2022] Open
Abstract
CD38 is a glycoprotein that is highly and uniformly expressed in plasma cells in multiple myeloma. A panel of CD38 and CD138/CD19/CD45/CD56/CD117 markers is considered the immunophenotypic diagnosis of plasma cell myeloma. Expression of the CD38 marker may fade or weaken compared with the CD138 marker in plasma cells after chemotherapy treatment. Herein we present a rare case of CD38-negative multiple myeloma that was initially misdiagnosed as acute leukemia.
Collapse
|
6
|
Liu Y, Ye X, Zhan X, Yu CY, Zhang J, Huang K. TPQCI: A topology potential-based method to quantify functional influence of copy number variations. Methods 2021; 192:46-56. [PMID: 33894380 DOI: 10.1016/j.ymeth.2021.04.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 12/21/2022] Open
Abstract
Copy number variation (CNV) is a major type of chromosomal structural variation that play important roles in many diseases including cancers. Due to genome instability, a large number of CNV events can be detected in diseases such as cancer. Therefore, it is important to identify the functionally important CNVs in diseases, which currently still poses a challenge in genomics. One of the critical steps to solve the problem is to define the influence of CNV. In this paper, we provide a topology potential based method, TPQCI, to quantify this kind of influence by integrating statistics, gene regulatory associations, and biological function information. We used this metric to detect functionally enriched genes on genomic segments with CNV in breast cancer and multiple myeloma and discovered biological functions influenced by CNV. Our results demonstrate that, by using our proposed TPQCI metric, we can detect disease-specific genes that are influenced by CNVs. Source codes of TPQCI are provided in Github (https://github.com/usos/TPQCI).
Collapse
Affiliation(s)
- Yusong Liu
- Collage of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin, Heilongjiang 150001, China; Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Xiufen Ye
- Collage of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin, Heilongjiang 150001, China
| | - Xiaohui Zhan
- Indiana University School of Medicine, Indianapolis, IN 46202, USA; National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518037, China; Department of Bioinformatics, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Christina Y Yu
- Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| | - Jie Zhang
- Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kun Huang
- Indiana University School of Medicine, Indianapolis, IN 46202, USA; Regenstrief Institute, Indianapolis, IN 46202, USA.
| |
Collapse
|
7
|
Sidana S, Jevremovic D, Ketterling RP, Tandon N, Dispenzieri A, Gertz MA, Greipp PT, Baughn LB, Buadi FK, Lacy MQ, Morice W, Hanson C, Timm M, Dingli D, Hayman SR, Gonsalves WI, Kapoor P, Kyle RA, Leung N, Go RS, Lust JA, Rajkumar SV, Kumar SK. Rapid assessment of hyperdiploidy in plasma cell disorders using a novel multi-parametric flow cytometry method. Am J Hematol 2019; 94:424-430. [PMID: 30592078 DOI: 10.1002/ajh.25391] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 12/18/2018] [Accepted: 12/26/2018] [Indexed: 12/23/2022]
Abstract
Trisomies of odd numbered chromosomes are seen in nearly half of patients with multiple myeloma (MM) and typically correlate with a hyperdiploid state and better overall survival (OS). We compared DNA ploidy of monoclonal plasma cells (as a surrogate for the presence of trisomies) assessed simultaneously by PCPRO (plasma cell proliferative index), a novel method that estimates DNA index by multi-parametric flow cytometry to fluorescence in situ hybridization (FISH) in 1703 patients with plasma cell disorders. The distribution of ploidy was hyperdiploid: 759 (45%), diploid 765 (45%), hypodiploid: 71 (4%), tetraploid/near-tetraploid: 108 (6%). FISH identified trisomies in 82% (621/756) of patients with hyperdiploidy by PCPRO and no trisomy by FISH was observed in 88% (730/834) of patients without hyperdiploidy. 95% (795/834) of patients without hyperdiploidy on PCPRO had one or less trisomy by FISH. Sensitivity and specificity of PCPRO for detecting hyperdiploidy was 86% (621/725) and 84% (730/865), respectively. Sensitivity increased to 94% (579/618) for patients with more than one trisomy. Newly diagnosed MM patients with hyperdiploidy on PCPRO (147/275) had better OS compared to nonhyperdiploid patients (median not reached vs 59 months, P = 0.008) and better progression free survival (median: 33 vs 23 months, P = 0.03). Within the hyperdiploidy group, patients with high-hyperdiploidy (DNA index: 1.19-1.50) versus those with low-hyperdiploidy (DNA index: 1.05-1.18) had superior OS (3 year OS of 88% vs 68% P = 0.03). Ploidy assessment by flow cytometry can provide rapid, valuable prognostic information and also reduces the number of copy number FISH probes required and hence the cost of FISH.
Collapse
Affiliation(s)
- Surbhi Sidana
- Division of Hematology Mayo Clinic Rochester Minnesota
| | - Dragan Jevremovic
- Department of Laboratory Medicine and Pathology Mayo Clinic Rochester Minnesota
| | - Rhett P. Ketterling
- Department of Laboratory Medicine and Pathology Mayo Clinic Rochester Minnesota
| | - Nidhi Tandon
- Division of Hematology Mayo Clinic Rochester Minnesota
| | | | | | - Patricia T Greipp
- Department of Laboratory Medicine and Pathology Mayo Clinic Rochester Minnesota
| | - Linda B. Baughn
- Division of Laboratory Genetics, Department of Laboratory Medicine and Pathology Mayo Clinic Rochester Minnesota
| | | | | | - William Morice
- Department of Laboratory Medicine and Pathology Mayo Clinic Rochester Minnesota
| | - Curtis Hanson
- Department of Laboratory Medicine and Pathology Mayo Clinic Rochester Minnesota
| | - Michael Timm
- Department of Laboratory Medicine and Pathology Mayo Clinic Rochester Minnesota
| | - David Dingli
- Division of Hematology Mayo Clinic Rochester Minnesota
| | | | | | | | | | - Nelson Leung
- Division of Hematology Mayo Clinic Rochester Minnesota
- Division of Nephrology Mayo Clinic Rochester Minnesota
| | - Ronald S. Go
- Division of Hematology Mayo Clinic Rochester Minnesota
| | - John A. Lust
- Division of Hematology Mayo Clinic Rochester Minnesota
| | | | | |
Collapse
|
8
|
Shah GL, Landau H, Londono D, Devlin SM, Kosuri S, Lesokhin AM, Lendvai N, Hassoun H, Chung DJ, Koehne G, Jhanwar SC, Landgren O, Levine R, Giralt SA. Gain of chromosome 1q portends worse prognosis in multiple myeloma despite novel agent-based induction regimens and autologous transplantation. Leuk Lymphoma 2017; 58:1823-1831. [PMID: 28078910 PMCID: PMC5554510 DOI: 10.1080/10428194.2016.1260126] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
We aimed to identify whether the use of autologous hematopoietic cell transplantation (HCT) impacts outcomes for multiple myeloma patients with gains of chromosome 1q (+1q). We retrospectively identified 95 patients, 21% having +1q. For patients with +1q, the overall response rate to induction was 85%, with 40% having ≥ VGPR and 20% achieving a CR, similar to non +1q patients (p = .64). The median PFS from diagnosis with +1q was 2.1 years (95% CI: 1.2-not reached (NR)) vs 4.3 years (95% CI: 3.3 yrs-NR) without +1q (p = .003). Median OS from diagnosis was 4.4 years (95% CI: 2.9-NR) vs not reached, respectively (p = .005). On molecular analysis using the Foundation One Heme assay, the most common mutations seen in +1q patients included TP53 (38%) and KRAS (25%). Overall, gain of 1q portends worse PFS and OS which was not negated by auto HCT. Such patients will likely require additional therapy to improve their survival.
Collapse
Affiliation(s)
- Gunjan L. Shah
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Heather Landau
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, NY Presbyterian Hospital – Weill Cornell Medical College, New York, NY, USA
| | - Dory Londono
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sean M. Devlin
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Satyajit Kosuri
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexander M. Lesokhin
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, NY Presbyterian Hospital – Weill Cornell Medical College, New York, NY, USA
| | - Nikoletta Lendvai
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, NY Presbyterian Hospital – Weill Cornell Medical College, New York, NY, USA
| | - Hani Hassoun
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, NY Presbyterian Hospital – Weill Cornell Medical College, New York, NY, USA
| | - David J. Chung
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, NY Presbyterian Hospital – Weill Cornell Medical College, New York, NY, USA
| | - Guenther Koehne
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, NY Presbyterian Hospital – Weill Cornell Medical College, New York, NY, USA
| | - Suresh C. Jhanwar
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ola Landgren
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, NY Presbyterian Hospital – Weill Cornell Medical College, New York, NY, USA
| | - Ross Levine
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, NY Presbyterian Hospital – Weill Cornell Medical College, New York, NY, USA
| | - Sergio A. Giralt
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, NY Presbyterian Hospital – Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
9
|
Wang D, Yu F, Fang B, Song Y. [Clinical analysis of four multiple myeloma patients with - 13/13q- and 17p]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2016; 37:65-7. [PMID: 26876257 PMCID: PMC7342308 DOI: 10.3760/cma.j.issn.0253-2727.2016.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
10
|
Lim AST, Krishnan S, Lim TH, See K, Ng YJ, Tan YM, Choo N, Lau LC, Tien SL, Ma J, Tan D. Amplification of 1q21 and other abnormalities in multiple myeloma patients from a tertiary hospital in singapore. Indian J Hematol Blood Transfus 2014; 30:253-8. [PMID: 25435723 DOI: 10.1007/s12288-013-0294-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 08/17/2013] [Indexed: 01/21/2023] Open
Abstract
Much effort has been made to stratify multiple myeloma patients for targeted therapy. However, responses have been varied and improved patient stratifications are needed. Forty-five diagnostic samples from multiple myeloma patients (median age 65 years) were stratified cytogenetically as 15 having non-hyperdiploidy, 20 having hyperdiploidy and 10 having a normal karyotype. Fluorescence in situ hybridization (FISH) assays with FGFR3/IGH, CCND1/IGH, IGH/MAF, RB1 and TP53 probes on bone marrow samples showed that IGH rearrangements were the most common abnormality in the non-hyperdiploid group but these were also found among hyperdiploid patients and patients with normal cytogenetics. Of these, FGFR3/IGH rearrangements were most frequent. Deletion of RB1/monosomy 13 was the most common genetic abnormality across the three groups and was significantly higher among non-hyperdiploid compared to hyperdiploid patients. On the other hand, the study recorded a low incidence of TP53 deletion/monosomy 17. The FGFR3/IGH fusion was frequently seen with RB1 deletion/monosomy 13. FISH with 1p36/1q21 and 6q21/15q22 probes showed that amplification of 15q22 was seen in all of the hyperdiploid patients while amplification of 1q21, Amp(1q21), characterized non-hyperdiploid patients. In contrast, deletions of 1p36 and 6q21 were very rare events. Amp(1q21), FGFR3/IGH fusion, RB1 deletion/monosomy 13, and even TP53 deletion/monosomy 17 were seen in some hyperdiploid patients, suggesting that they have a less than favorable prognosis and require closer monitoring.
Collapse
Affiliation(s)
- Alvin S T Lim
- Cytogenetics Laboratory, Department of Pathology, Singapore General Hospital, The Academia-Diagnostics Tower, L9, 20 College Road, Singapore, 169856 Singapore
| | - Sathish Krishnan
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Tse Hui Lim
- Cytogenetics Laboratory, Department of Pathology, Singapore General Hospital, The Academia-Diagnostics Tower, L9, 20 College Road, Singapore, 169856 Singapore
| | - Karen See
- Cytogenetics Laboratory, Department of Pathology, Singapore General Hospital, The Academia-Diagnostics Tower, L9, 20 College Road, Singapore, 169856 Singapore
| | - Yit Jun Ng
- Cytogenetics Laboratory, Department of Pathology, Singapore General Hospital, The Academia-Diagnostics Tower, L9, 20 College Road, Singapore, 169856 Singapore
| | - Yu Min Tan
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Natasha Choo
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Lai Ching Lau
- Cytogenetics Laboratory, Department of Pathology, Singapore General Hospital, The Academia-Diagnostics Tower, L9, 20 College Road, Singapore, 169856 Singapore
| | - Sim Leng Tien
- Cytogenetics Laboratory, Department of Pathology, Singapore General Hospital, The Academia-Diagnostics Tower, L9, 20 College Road, Singapore, 169856 Singapore ; Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Jun Ma
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC Australia
| | - Daryl Tan
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
11
|
Lee JW, Lee JK, Hong YJ, Hong SI, Chang YH. Correlation of Chromosomal Aberrations with Prognostic Markers in Multiple Myeloma Patients-A Single Institution Study. Ann Lab Med 2008; 28:413-8. [DOI: 10.3343/kjlm.2008.28.6.413] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Ji Won Lee
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Seoul, Korea
| | - Jin Kyung Lee
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Seoul, Korea
| | - Young Joon Hong
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Seoul, Korea
| | - Seok-Il Hong
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Seoul, Korea
| | - Yoon Hwan Chang
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Seoul, Korea
| |
Collapse
|
12
|
Leone PE, Walker BA, Jenner MW, Chiecchio L, Dagrada G, Protheroe RKM, Johnson DC, Dickens NJ, Brito JL, Else M, Gonzalez D, Ross FM, Chen-Kiang S, Davies FE, Morgan GJ. Deletions of CDKN2C in multiple myeloma: biological and clinical implications. Clin Cancer Res 2008; 14:6033-41. [PMID: 18829482 DOI: 10.1158/1078-0432.ccr-08-0347] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
PURPOSE Deletions of chromosome 1 have been described in 7% to 40% of cases of myeloma with inconsistent clinical consequences. CDKN2C at 1p32.3 has been identified in myeloma cell lines as the potential target of the deletion. We tested the clinical impact of 1p deletion and used high-resolution techniques to define the role of CDKN2C in primary patient material. EXPERIMENTAL DESIGN We analyzed 515 cases of monoclonal gammopathy of undetermined significance (MGUS), smoldering multiple myeloma (SMM), and newly diagnosed multiple myeloma using fluorescence in situ hybridization (FISH) for deletions of CDKN2C. In 78 myeloma cases, we carried out Affymetrix single nucleotide polymorphism mapping and U133 Plus 2.0 expression arrays. In addition, we did mutation, methylation, and Western blotting analysis. RESULTS By FISH we identified deletion of 1p32.3 (CDKN2C) in 3 of 66 MGUS (4.5%), 4 of 39 SMM (10.3%), and 55 of 369 multiple myeloma cases (15%). We examined the impact of copy number change at CDKN2C on overall survival (OS), and found that the cases with either hemizygous or homozygous deletion of CDKN2C had a worse OS compared with cases that were intact at this region (22 months versus 38 months; P = 0.003). Using gene mapping we identified three homozygous deletions at 1p32.3, containing CDKN2C, all of which lacked expression of CDKN2C. Cases with homozygous deletions of CDKN2C were the most proliferative myelomas, defined by an expression-based proliferation index, consistent with its biological function as a cyclin-dependent kinase inhibitor. CONCLUSIONS Our results suggest that deletions of CDKN2C are important in the progression and clinical outcome of myeloma.
Collapse
Affiliation(s)
- Paola E Leone
- Section of Haemato-Oncology, The Institute of Cancer Research, 15 Cotswold Road, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Sreekantaiah C. FISH panels for hematologic malignancies. Cytogenet Genome Res 2007; 118:284-96. [DOI: 10.1159/000108312] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2007] [Accepted: 06/18/2007] [Indexed: 11/19/2022] Open
|
14
|
Fournier A, Florin A, Lefebvre C, Solly F, Leroux D, Callanan M. Genetics and epigenetics of 1q rearrangements in hematological malignancies. Cytogenet Genome Res 2007; 118:320-7. [DOI: 10.1159/000108316] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Accepted: 02/09/2007] [Indexed: 12/11/2022] Open
|
15
|
Moon HW, Cho HI, Kang SH, Kim TY, Min HC, Oh BR, Yoon SS, Lee DS. IgH gene rearrangements are associated with hyperdiploid multiple myeloma in Korean cases: a fluorescence in situ hybridization study. CANCER GENETICS AND CYTOGENETICS 2007; 172:172-3. [PMID: 17213029 DOI: 10.1016/j.cancergencyto.2006.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2006] [Accepted: 08/24/2006] [Indexed: 05/13/2023]
|
16
|
Fonseca R, Van Wier SA, Chng WJ, Ketterling R, Lacy MQ, Dispenzieri A, Bergsagel PL, Rajkumar SV, Greipp PR, Litzow MR, Price-Troska T, Henderson KJ, Ahmann GJ, Gertz MA. Prognostic value of chromosome 1q21 gain by fluorescent in situ hybridization and increase CKS1B expression in myeloma. Leukemia 2006; 20:2034-40. [PMID: 17024118 DOI: 10.1038/sj.leu.2404403] [Citation(s) in RCA: 158] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A specific role for increased level of expression of CKS1B, as a consequence of chromosome 1q21 copy number gain, has been postulated as both pathogenic, as well as a powerful clinical prognostic factor in multiple myeloma (MM). The purpose of this study is to determine the clinical associations and prognostic impact of copy number gain at chromosome 1q21 (with a bacteria artificial chromosome clone containing CKS1B) and CKS1B gene level of expression in MM. We studied the chromosome region 1q21 for copy number change in a cohort of myeloma patients treated by high-dose therapy with stem-cell rescue (HDT) (n = 159). A separate cohort of patients, treated by HDT was studied for CKS1B messenger RNA expression by gene expression profiling (n = 67). 1q21 gain was then correlated with clinical parameters and survival. Gain of 1q21 copy number was detected in about a third of MM and was associated with more proliferative disease and poor-risk cytogenetic categories such as t(4;14), and chromosome 13 deletion. Both 1q21 gain and increase gene expression level were significantly associated with reduced survival. However, neither is an independent prognostic marker in MM on multivariate Cox proportional hazard analysis.
Collapse
Affiliation(s)
- R Fonseca
- Division of Hematology and Oncology, Mayo Clinic Comprehensive Cancer Center, Scottsdale, AZ 85259, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Liebisch P, Döhner H. Cytogenetics and molecular cytogenetics in multiple myeloma. Eur J Cancer 2006; 42:1520-9. [PMID: 16781866 DOI: 10.1016/j.ejca.2005.12.028] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2005] [Accepted: 12/20/2005] [Indexed: 12/22/2022]
Abstract
Multiple myeloma (MM) is characterized by frequent and complex genomic abnormalities that not only essentially contribute to the pathogenesis of this disease but also reflect its prognostic heterogeneity. There is evidence for two more or less mutually exclusive oncogenic pathways in the early development of clonal plasma cell disorders. Approximately half the tumours are non-hyperdiploid and carry translocations of the immunoglobulin heavy-chain (IgH) locus and various oncogenes, for example Cyclin D1, Cyclin D3, and FGFR3. The remaining hyperdiploid tumours exhibit recurrent trisomies - typically of chromosomes 5, 7, 9, 11, 15, 19, and 21 - but infrequently exhibit IgH translocations. While some chromosomal aberrations, such as deletion of chromosome arm 13q, deliver independent prognostic information that is already utilized for risk stratification within clinical trials, the prognostic significance of most other genetic aberrations in MM is undetermined.
Collapse
Affiliation(s)
- Peter Liebisch
- Department of Internal Medicine III, University Hospital of Ulm, Robert-koch-str. 8, 89081 Ulm, Germany.
| | | |
Collapse
|
18
|
Zhan F, Huang Y, Colla S, Stewart JP, Hanamura I, Gupta S, Epstein J, Yaccoby S, Sawyer J, Burington B, Anaissie E, Hollmig K, Pineda-Roman M, Tricot G, van Rhee F, Walker R, Zangari M, Crowley J, Barlogie B, Shaughnessy JD. The molecular classification of multiple myeloma. Blood 2006; 108:2020-8. [PMID: 16728703 PMCID: PMC1895543 DOI: 10.1182/blood-2005-11-013458] [Citation(s) in RCA: 854] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
To better define the molecular basis of multiple myeloma (MM), we performed unsupervised hierarchic clustering of mRNA expression profiles in CD138-enriched plasma cells from 414 newly diagnosed patients who went on to receive high-dose therapy and tandem stem cell transplants. Seven disease subtypes were validated that were strongly influenced by known genetic lesions, such as c-MAF- and MAFB-, CCND1- and CCND3-, and MMSET-activating translocations and hyperdiploidy. Indicative of the deregulation of common pathways by gene orthologs, common gene signatures were observed in cases with c-MAF and MAFB activation and CCND1 and CCND3 activation, the latter consisting of 2 subgroups, one characterized by expression of the early B-cell markers CD20 and PAX5. A low incidence of focal bone disease distinguished one and increased expression of proliferation-associated genes of another novel subgroup. Comprising varying fractions of each of the other 6 subgroups, the proliferation subgroup dominated at relapse, suggesting that this signature is linked to disease progression. Proliferation and MMSET-spike groups were characterized by significant overexpression of genes mapping to chromosome 1q, and both exhibited a poor prognosis relative to the other groups. A subset of cases with a predominating myeloid gene expression signature, excluded from the profiling analyses, had more favorable baseline characteristics and superior prognosis to those lacking this signature.
Collapse
Affiliation(s)
- Fenghuang Zhan
- Donna D. and Donald M. Lambert Laboratory of Myeloma Genetics, Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Hanamura I, Stewart JP, Huang Y, Zhan F, Santra M, Sawyer JR, Hollmig K, Zangarri M, Pineda-Roman M, van Rhee F, Cavallo F, Burington B, Crowley J, Tricot G, Barlogie B, Shaughnessy JD. Frequent gain of chromosome band 1q21 in plasma-cell dyscrasias detected by fluorescence in situ hybridization: incidence increases from MGUS to relapsed myeloma and is related to prognosis and disease progression following tandem stem-cell transplantation. Blood 2006; 108:1724-32. [PMID: 16705089 PMCID: PMC1895503 DOI: 10.1182/blood-2006-03-009910] [Citation(s) in RCA: 352] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Using fluorescence in situ hybridization we investigated amplification of chromosome band 1q21 (Amp1q21) in more than 500 untreated patients with monoclonal gammopathy of undetermined significance (MGUS; n = 14), smoldering multiple myeloma (SMM; n = 31), and newly diagnosed MM (n = 479) as well as 45 with relapsed MM. The frequency of Amp1q21 was 0% in MGUS, 45% in SMM, 43% in newly diagnosed MM, and 72% in relapsed MM (newly diagnosed versus relapsed MM, P < .001). Amp1q21 was detected in 10 of 12 patients whose disease evolved to active MM compared with 4 of 19 who remained with SMM (P < .001). Patients with newly diagnosed MM with Amp1q21 had inferior 5-year event-free/overall survival compared with those lacking Amp1q21 (38%/52% versus 62%/78%, both P < .001). Thalidomide improved 5-year EFS in patients lacking Amp1q21 but not in those with Amp1q21 (P = .004). Multivariate analysis including other major predictors revealed that Amp1q21 was an independent poor prognostic factor. Relapsed patients who had Amp1q21 at relapse had inferior 5-year postrelapse survival compared with those lacking Amp1q21 at relapse (15% versus 53%, P = .027). The proportion of cells with Amp1q21 and the copy number of 1q21 tended to increase at relapse compared with diagnosis. Our data suggest that Amp1q21 is associated with both disease progression and poor prognosis.
Collapse
Affiliation(s)
- Ichiro Hanamura
- Donna D. and Donald M. Lambert Laboratory of Myeloma Genetics, Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, 4301 W. Markham St. #776, Little Rock, AR 72205, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Cremer FW, Kartal M, Hose D, Bila J, Buck I, Bellos F, Raab MS, Brough M, Moebus A, Hager HD, Goldschmidt H, Moos M, Bartram CR, Jauch A. High incidence and intraclonal heterogeneity of chromosome 11 aberrations in patients with newly diagnosed multiple myeloma detected by multiprobe interphase FISH. ACTA ACUST UNITED AC 2005; 161:116-24. [PMID: 16102581 DOI: 10.1016/j.cancergencyto.2005.02.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2004] [Revised: 02/10/2005] [Accepted: 02/15/2005] [Indexed: 01/14/2023]
Abstract
In multiple myeloma, additional copies of chromosome 11 material, reported to confer an unfavorable prognosis, have been found in 20-45% of patients. To assess the incidence and extent of chromosome 11 aberrations, we performed interphase fluorescence in situ hybridization on CD138+ bone marrow plasma cells of 50 newly diagnosed myeloma patients, using seven locus-specific probes for chromosome 11, one for 13q14.3, and a probe set for translocation t(11;14). In 33 of 50 patients, chromosome 11 aberrations were found. Results indicated a marked intraclonal heterogeneity: in 13 patients, trisomy 11; in 10 patients, subclones with trisomy 11 and partial trisomies 11q coexisted; in 6 patients, only a partial trisomy 11q; and in 6 patients, a tetrasomy or partial tetrasomy 11. The coexistence of subclones with varying extent and copy numbers of chromosome 11 material indicates ongoing structural changes and clonal evolution. Hybridization results delineated 11q23 and 11q25 as the most frequently gained regions, which supports a relevant pathogenetic role of genes on 11q23 and 11q25. To confirm the high incidence of 11q23 gains, a further 50 patients (total n=100) were analyzed for 11q23 and 13q14.3. Myeloma with gains of 11q23 showed a low frequency of deletion 13q14.3 and may prove to be a distinct subgroup of this disease.
Collapse
Affiliation(s)
- Friedrich W Cremer
- Institute of Human Genetics, University of Heidelberg, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Liebisch P, Scheck D, Erné SA, Wellmann A, Wendl C, Janczik S, Kolmus S, Kröber A, Einsele H, Straka C, Goldschmidt H, Benner A, Stilgenbauer S, Döhner H. Duplication of chromosome arms 9q and 11q: evidence for a novel, 14q32 translocation-independent pathogenetic pathway in multiple myeloma. Genes Chromosomes Cancer 2005; 42:78-81. [PMID: 15390182 DOI: 10.1002/gcc.20098] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
14q32 translocations [t(14q)] represent critical but not universal events in multiple myeloma (MM). Gains of chromosome arms 1q, 9q, and 11q (+1q, +9q, and +11q) have recently been identified as frequent aberrations in this disease, but their pathogenetic significance remains unclear. We studied a series of 108 MM patients using fluorescence in situ hybridization and DNA probes mapping to chromosome bands 1q21, 9q34, 11q25, 13q14, and 14q32. Three subsets of tumors were defined: (1) MM+/+ (detection of +9q and +11q; 43.5% of cases), (2) MM+/- (+9q or +11q; 21.3%), and (3) MM-/- (neither +9q nor +11q; 35.2%). The incidence of t(14q) was significantly different in these subgroups: 23% in MM+/+, 56% in MM+/-, and 89% in MM-/-. Deletion of 13q (13q-) also was significantly less frequent in MM+/+ (23%) than in MM+/- and MM-/- (36% and 63%, respectively). The nonrandom distribution of chromosomal aberrations in the present series of MM tumors points to a novel, 14q32 translocation-independent pathogenetic pathway in plasma cell neoplasms.
Collapse
Affiliation(s)
- Peter Liebisch
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Cremer FW, Bila J, Buck I, Kartal M, Hose D, Ittrich C, Benner A, Raab MS, Theil AC, Moos M, Goldschmidt H, Bartram CR, Jauch A. Delineation of distinct subgroups of multiple myeloma and a model for clonal evolution based on interphase cytogenetics. Genes Chromosomes Cancer 2005; 44:194-203. [PMID: 16001433 DOI: 10.1002/gcc.20231] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
To delineate multiple myeloma (MM) subgroups and their clonal evolution, we analyzed 81 newly diagnosed patients by interphase fluorescence in situ hybridization using a comprehensive probe set for 10 chromosomes and two IGH rearrangements. A median of 5 probes per patient displayed aberrant signal numbers (range, 1-10). Additional copies most frequently found were for 15q22, 19q13, 9q34, 11q23, and 1q21. Losses commonly observed were of 13q14.3, 17p13, and 22q11. Predominance of gain or loss was quantified by a copy number score (CS) for each patient. Two peaks (CS = +3 and CS = 0) were found by plotting patient copy number scores over CS values corresponding to hyperdiploid and nonhyperdiploid MM. Cluster analysis revealed four major branches: (i) gain of 9q, 15q, 19q, and/or 11q; (ii) deletion of 13q and t(4;14); (iii) t(11;14); and (iv) gain of 1q. Statistical modeling of an oncogenetic tree indicated that early independent events were gain of 15q/9q and/or 11q, t(11;14); deletion of 13q followed by t(4;14); and gain of 1q. Aberrations of 17p13, 22q11, 8p12, and 6q21 were found as subsequent events. MM with gain of 1q was delineated as a subentity with significantly higher beta-2-microglobulin and lower hemoglobin levels, indicating a poor prognosis. From our results, we propose a model of MM for clonal evolution.
Collapse
|