Molecular Cytogenetics of Multiple Myeloma

Research Article

Molecular Cytogenetics of Multiple Myeloma

Gozzetti A*, Frasconi A and Crupi R

Division of Hematology and Transplants, University of Siena, Italy

*Corresponding author: Alessandro Gozzetti, Division of Hematology and Transplants, University of Siena, Policlinico “Santa Maria Le Scotte” 53100 Siena, Italy.

Received: July 28, 2014; Accepted: November 24, 2014; Published: December 01, 2014

Citation: Gozzetti A, Frasconi A and Crupi R. Molecular Cytogenetics of Multiple Myeloma. Austin J Cancer Clin Res 2014;1(4): 1020. ISSN 2381-909X


Abstract

Multiple Myeloma (MM) is a plasma cell tumor characterized by multiple genetic lesions. Two main groups of abnormalities can be distinguished: Hyper diploid and hipodoploid, in which the chromosome number is mainly constituted by trisomies or monosomies, respectively. The advent of Fluorescence In Situ Hybridization (FISH) has permitted to identify genomic lesions that have been integrated into a prognostic score system that take in consideration also clinical parameters. This simple analysis can be part of the initial evaluation of the patient and can drive more accurately his treatment.

Keywords: Myeloma; Cytogenetics; Fish

Introduction

In contrast to other haematological neoplasms such as the acute leukaemias, the systematic investigation of chromosomal aberrations in Multiple Myeloma (MM) by the use of conventional cytogenetics has been hampered by the low mitotic activity of tumour cells in this disease [1,2]. With the introduction of molecular-based cytogenetic techniques into the analysis of MM, and its precursor condition Monoclonal Gammopathy of Undetermined Significance (MGUS), considerable advances in the understanding of the biology of plasma cell tumours has been achieved. Using Fluorescence In Situ Hybridization (FISH), multiple and complex chromosomal abnormalities are detectable in virtually all patients with MM and most patients with MGUS with great prognostic importance [3].

Conventional cytogenetics

Cytogenetic analysis in MM is informative in less than 30% of patients this is due to the mitotic activity of tumour cells that in MM as compared to other haematological diseases is generally very low. Karyotypes are typically complex and exhibit more than 10 abnormalities in almost half of patients [4].

Numerical changes

The numerical changes are monosomies of chromosomes 13, 14, 16, and 22 as well as trisomies of chromosomes 3, 5, 7, 9, 11, 15, 19, and 21 [5]. Independent of the detection of specific chromosomal aberrations, chromosome banding analysis provides valuable prognostic information that can firstly be extracted from the presence or absence of abnormal metaphases, and secondly from the tumour cell ploidy in informative cases [6]. Patients with a normal karyotype enjoy a significantly longer survival than those that are cytogenetically abnormal [7]. Moreover, the classification of tumours with aberrant metaphases according to their chromosome number is also of prognostic relevance. In several series, hypodiploidy was associated with a significantly inferior outcome. However, as other adverse genetic features – such as monosomy 13/13q deletion, t(4;14), and t(14;16) – are predominantly present in hypodiploid tumours, it remains controversial whether or not hypodiploidy is an independent prognostic marker [8-10]. While numerical changes can be diagnosed easily, small interstitial deletions are usually missed. Fluorescence In Situ Hybridization (FISH), allow the detection of genetic abnormalities independently of proliferating cells [3,11]. With these methods, chromosomal aberrations are found in virtually all patients with MM. It is very important that plasmacells are purified from the original sample [2]. This is due to variable infiltration of plasmacells in the bone marrow and the dilution in the bone marrow aspirate that can lead to false negative results. There are various techniques to enrich tumour cells in MM. The most widely used method applies magnetic microbeads that recognize the cell surface antigen CD138 expressed on both, normal as well as malignant plasma cells fish [12]. FISH permits the reliable identification of translocations as well as small deletions or gains. FISH became the most widely used technique for the analysis of chromosomal abnormalities in MM.

Fish

The indication in MM is to test for 13q14 and 17p deletions (13q-, 17p-), translocations of the IGH locus, in particular t(4;14) (p16.3;q32) , t(11;14)(q13;q32) , t(14;16) (q32; q23) and abnormalities of chromosome 1p and 1q [2]. Most of these probes are available. For the detection of 13q loss, most laboratories apply probes mapping to chromosome band 13q14, although the critical region of 13q- is still poorly defined. For the detection of 17p-, it is general practice to apply probes containing the p53 gene. Translocations involving the immunoglobulin heavy-chain (IgH) locus can be detected by the use of DNA probes mapping to the constant (CH) and variable region of the IgH gene [13]. Reciprocal translocations are diagnosed by the colocalization of differentially labelled probes for IgH and the respective translocation partner (e.g. 11q13, 4p16, 16q23), ideally on both derivative chromosomes (double fusion). Cut-off levels for positive results are based on data obtained from bone marrow specimens of healthy volunteers. For dual fusion or break-apart probes, a cut-off level of 10% could be accepted. For single fusion results with dual fusion probes, a cut-off level of 20% can be good, although some laboratories apply a threshold of 10% for these probes, and it is important that each laboratory set proper cut off values [2].

13q deletion

By FISH in several studies, -13/13q- is present in about 50% of cases representing the most frequent abnormality in MM [14]. Using conventional cytogenetics, the incidence of chromosome 13 losses among patients with informative karyotypes is comparable, resulting in an overall incidence of 10% to 20% as shown in large cytogenetic series. In patients with t(4;14)(p16.3;q32) or t(14;16)(q32;q23), the incidence of -13/13q- is approximately 90%. By conventional cytogenetics -13/13q- is associated with significantly lower response rates, shorter Event-Free Survival (EFS), and inferior Overall Survival (OS) in MM. This is true for patients after conventional chemotherapy as well as for patients treated with High-Dose Chemotherapy (HD-CTX) and Autologous Stem Cell Transplantation (ASCT) [15]. -13/13q- by karyotyping predict a more unfavourable prognosis than the detection of the same abnormality by FISH. This is most likely due to a combination of negative prognostic markers reflected by this finding (higher rate of proliferating cells) [16]. Moreover, significantly more 13q-deleted patients are identified by FISH as compared to conventional cytogenetics (40–50% vs. 15–20%).

17p deletion

Inactivation of the p53 tumour suppressor gene by monoallelic deletion or mutation is associated with disease progression in many human malignancies. In most FISH series, the incidence of p53 deletion among newly diagnosed patients was in the range of 5% to 10% [17,18]. However, functional loss of the gene is present in up to 40% of patients with advanced MM and in more than 60% of human myeloma cell lines, pointing to this abnormality as a marker of tumour progression [19]. Independent of the mode of treatment (conventional chemotherapy or HD-CTX), deletion of the p53 gene locus identified by FISH is a predictor of shorter survival [20].

Chromosome 1q

In cytogenetic studies, 1q abnormalities were associated with advanced disease and tumour progression, as well as with shorter event-free survival. The gene implicated seems amplificated and is the cell cycle regulator gene CKS1B at chromosome band 1q21 as a predictor of a particularly unfavourable prognosis [21-23].

IgH translocations

IgH translocations are equally frequent in, MGUS and MM (~40– 60%), which strongly suggests that primary IgH rearrangements represent early pathogenetic events [24]. The overall rate of 14q32 translocations, however, significantly increases with disease progression and reaches up to 90% in advanced tumours and Human Myeloma Cell Lines (HMCL), most likely reflecting a rising number of secondary IgH translocations which seem to be virtually absent in MGUS and smoldering MM. Typically, there are five main translocations involving 11q13 (CCND1), 6p21 (CCND3), 16q23 (MAF), 20q12 (MAFB), and 4p16 (FGFR3 and MMSET) [25].

t(11;14)(q13;q32)

Tumours carrying the t(11;14)(q13;q32) can be identified either by chromosome banding analysis, FISH, or gene expression analysis. Using FISH, the rearrangement can be identified in about 15–20% of patients with MM and 15–30% of cases with MGUS [26,27]. The presence of t(11;14) has been correlated with a lymphoplasmacytic, mature morphology of plasma cells, CD20 expression, and the oligo-/ asecretory MM subtype. There was no impact of t(11;14) on survival of patients treated with conventional chemotherapy. The t(11;14) (q13;q32) is identical to that observed in mantle-cell lymphomas. The breakpoints involve the IGH gene at 14q32, and the CCND1 gene at 11q13, encoding the cyclin D1 protein. Although the cyclin D1 has been involved in the activation of proliferation, the t(11;14) myelomas are characterized by a low proliferative index and a frequent morphology of small mature plasma cells. Clinically, this type of myeloma is not remarkable. Although preliminary reports did show a better survival, more recent and larger studies did not confirm this prognostic impact [28].

t(4;14)(p16.3;q32)

The karyotypically cryptic t(4;14)(p16.3;q32) is detectable in approximately 15–20% of primary tumour specimens by FISH and leads to the dysregulation of two oncogenes, MMSET on der(4) and FGFR3 on der(14) [21,29,30]. Of note, in about 20% of cases with t(4;14), FGFR3 on der(14) is lost or not expressed. The translocation was found to be more prevalent among tumours with an IgA isotype as well as in patients with aggressive clinical features. Independently from the mode of treatment, t(4;14) is associated with an unfavourable clinical course [31].

t(14;16)(q32;q23)

Like t(4;14), t(14;16)(q32;q23) is karyotypically silent but can reliably be identified by FISH. With an incidence of 2–10%, t(14;16) is comparatively rare. t(14;16) results in the upregulation of the basic leucine zipper (bZIP) transcription factor c-maf. There is only scarce data about the prognostic impact of this IgH rearrangement, but it seems associated with poor prognosis [32,33].

Conclusion

The analysis of genomic rearrangements in MGUS and MM by the use of cytogenetics and molecular cytogenetics has ameliorated our understanding of clonal plasma cell disorders. The evaluation of a comprehensive panel of chromosomal imbalances and translocations is currently important within clinical management.

References

  1. Fonseca R, San Miguel J. Prognostic factors and staging in multiple myeloma. Hematol Oncol Clin North Am. 2007; 21: 1115-1140.
  2. Ross FM, Avet-Loiseau H, Ameye G, Gutiérrez NC, Liebisch P, O'Connor S, et al. Report from the European Myeloma Network on interphase FISH in multiple myeloma and related disorders. Haematologica. 2012; 97: 1272-1277.
  3. Gozzetti A, Le Beau MM. Fluorescence in situ hybridization: uses and limitations. Semin Hematol. 2000; 37: 320-333.
  4. Sawyer JR. The prognostic significance of cytogenetics and molecular profiling in multiple myeloma. Cancer Genet. 2011; 204: 3-12.
  5. Avet-Loiseau H. Role of genetics in prognostication in myeloma. Best Pract Res Clin Haematol. 2007; 20: 625-635.
  6. Fonseca R, Barlogie B, Bataille R, Bastard C, Bergsagel PL, Chesi M, et al. Genetics and cytogenetics of multiple myeloma: a workshop report. Cancer Res. 2004; 64: 1546-1558.
  7. Barlogie B, Shaughnessy JD. Early results of total therapy II in multiple myeloma: implications of cytogenetics and FISH. Int J Hematol. 2002; 76: 337-339.
  8. Higgins MJ, Fonseca R. Genetics of multiple myeloma. Best Pract Res Clin Haematol. 2005; 18: 525-536.
  9. Cigudosa JC, Calasanz MJ, Odero MD, Prosper F, Etxaniz A, Marin J, et al. Cytogenetic data in 41 patients with multiple myeloma. Karyotype and other clinical parameters. Cancer Genet Cytogenet. 1994; 78: 210-213.
  10. Dewald GW, Therneau T, Larson D, Lee YK, Fink S, Smoley S, et al. Relationship of patient survival and chromosome anomalies detected in metaphase and/or interphase cells at diagnosis of myeloma. Blood. 2005; 106: 3553-3558.
  11. Avet-Loiseau H, Durie BG, Cavo M, Attal M, Gutierrez N, Haessler J, et al. Combining fluorescent in situ hybridization data with ISS staging improves risk assessment in myeloma: an International Myeloma Working Group collaborative project. Leukemia. 2013; 27: 711-717.
  12. Hose D, Seckinger A, Jauch A, Rème T, Moreaux J, Bertsch U, et al. The role of fluorescence in situ hybridization and gene expression profiling in myeloma risk stratification. Srp Arh Celok Lek. 2011; 139: 84-89.
  13. Bergsagel PL, Kuehl WM. Chromosome translocations in multiple myeloma. Oncogene. 2001; 20: 5611-5622.
  14. Avet-Louseau H, Daviet A, Sauner S, Bataille R. Intergroupe Francophone du Myélome. Chromosome 13 abnormalities in multiple myeloma are mostly monosomy 13. Br J Haematol. 2000; 111: 1116-1117.
  15. Becze E. Cytogenetics helps determine diagnosis and prognosis for multiple myeloma. ONS Connect. 2012; 27: 18-19.
  16. Chiecchio L, Protheroe RK, Ibrahim AH, Cheung KL, Rudduck C, Dagrada GP, et al. Deletion of chromosome 13 detected by conventional cytogenetics is a critical prognostic factor in myeloma. Leukemia. 2006; 20: 1610-1617.
  17. Avet-Loiseau H, Li JY, Godon C, Morineau N, Daviet A, Harousseau JL, et al. P53 deletion is not a frequent event in multiple myeloma. Br J Haematol. 1999; 106: 717-719.
  18. Chng WJ, Price-Troska T, Gonzalez-Paz N, Van Wier S, Jacobus S, Blood E, et al. Clinical significance of TP53 mutation in myeloma. Leukemia. 2007; 21: 582-584.
  19. Lodé L, Eveillard M, Trichet V, Soussi T, Wuillème S, Richebourg S, et al. Mutations in TP53 are exclusively associated with del(17p) in multiple myeloma. Haematologica. 2010; 95: 1973-1976.
  20. López-Anglada L, Gutiérrez NC, García JL, Mateos MV, Flores T, San Miguel JF. P53 deletion may drive the clinical evolution and treatment response in multiple myeloma. Eur J Haematol. 2010; 84: 359-361.
  21. Avet-Loiseau H, Attal M, Campion L, Caillot D, Hulin C, Marit G, et al. Long-term analysis of the IFM 99 trials for myeloma: cytogenetic abnormalities [t(4;14), del(17p), 1q gains] play a major role in defining long-term survival. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2012; 30:1949-1952.
  22. Chang H, Qi X, Trieu Y, Xu W, Reader JC, Ning Y, et al. Multiple myeloma patients with CKS1B gene amplification have a shorter progression-free survival post-autologous stem cell transplantation. Br J Haematol. 2006; 135: 486-491.
  23. Marzin Y, Jamet D, Douet-Guilbert N, Morel F, Le Bris MJ, Morice P, et al. Chromosome 1 abnormalities in multiple myeloma. Anticancer Res. 2006; 26: 953-959.
  24. Fonseca R, Debes-Marun CS, Picken EB, Dewald GW, Bryant SC, Winkler JM, et al. The recurrent IgH translocations are highly associated with nonhyperdiploid variant multiple myeloma. Blood. 2003; 102: 2562-2567.
  25. Avet-Loiseau H, Facon T, Daviet A, Godon C, Rapp MJ, Harousseau JL, et al. 14q32 translocations and monosomy 13 observed in monoclonal gammopathy of undetermined significance delineate a multistep process for the oncogenesis of multiple myeloma. Intergroupe Francophone du Myelome. Cancer research. 1999; 59: 4546-4550.
  26. An G, Xu Y, Shi L, Zou D, Deng S, Sui W, et al. t(11;14) multiple myeloma: a subtype associated with distinct immunological features, immunophenotypic characteristics but divergent outcome. Leukemia research. 2013; 37: 1251-1257.
  27. Avet-Loiseau H, Garand R, Lodé L, Harousseau JL, Bataille R. Intergroupe Francophone du Myélome . Translocation t(11;14)(q13;q32) is the hallmark of IgM, IgE, and nonsecretory multiple myeloma variants. Blood. 2003; 101: 1570-1571.
  28. Fonseca R, Blood EA, Oken MM, Kyle RA, Dewald GW, Bailey RJ, et al. Myeloma and the t(11;14)(q13;q32); evidence for a biologically defined unique subset of patients. Blood. 2002; 99: 3735-3741.
  29. Avet-Loiseau H, Brigaudeau C, Morineau N, Talmant P, Lai JL, Daviet A, et al. High incidence of cryptic translocations involving the Ig heavy chain gene in multiple myeloma, as shown by fluorescence in situ hybridization. Genes, chromosomes & cancer. 1999; 24: 9-15.
  30. Chang H, Sloan S, Li D, Zhuang L, Yi QL, Chen CI, et al. The t(4;14) is associated with poor prognosis in myeloma patients undergoing autologous stem cell transplant. Br J Haematol. 2004; 125: 64-68.
  31. Winkler JM, Greipp P, Fonseca R. t(4;14)(p16.3;q32) is strongly associated with a shorter survival in myeloma patients. Br J Haematol. 2003; 120: 170-171.
  32. Avet-Loiseau H, Malard F, Campion L, Magrangeas F, Sebban C, Lioure B, et al. Translocation t(14;16) and multiple myeloma: is it really an independent prognostic factor? Blood. 2011; 117: 2009-2011.
  33. Rajkumar SV. Multiple myeloma: 2013 update on diagnosis, risk-stratification, and management. Am J Hematol. 2013; 88: 226-235.

Copyright © 2014 Journal.com. All Rights Reserved.