CC BY 4.0 · Indian J Med Paediatr Oncol 2023; 44(05): 505-514
DOI: 10.1055/s-0043-1762920
Review Article

Role of Conventional Cytogenetics and FISH in the Laboratory Work Up of Plasma Cell Dyscrasias

Aaishwarya Dhabe
1   Department of Cytogenetics, Tata Medical Center, MAR(EW), Newtown, Kolkata, India
,
Samipa Das
1   Department of Cytogenetics, Tata Medical Center, MAR(EW), Newtown, Kolkata, India
,
Mayur Parihar
1   Department of Cytogenetics, Tata Medical Center, MAR(EW), Newtown, Kolkata, India
› Author Affiliations

Abstract

Plasma cell dyscrasias are a heterogeneous group of neoplasms characterized by abnormal proliferation of plasma cells with or without over production of monoclonal immunoglobulins. Chromosomal abnormalities are acquired either early in the course of the disease or during disease progression. Plasma cell dyscrasias are categorized into multiple cytogenetic subtypes that form an integral component of risk-stratified treatment protocols. The primary genetic events are IgH gene translocations and non-random gains of chromosomes 3/5/7/9/11/15/19 and or 21. The secondary genetic events consist of chromosome 1 abnormalities (1p deletion and 1q gain or amplification), deletion 17p/TP53, deletion 13q, and MYC gene rearrangements. Plasma cells being at the end of differentiation spectrum of B cells, have low proliferative potential precluding the use of karyotyping in identification of chromosomal abnormalities. Analysis of enriched plasma cells using interphase fluorescent in situ hybridization (FISH) is the technique of choice for identifying these abnormalities. It is essential to enrich plasma cells before the FISH analysis, and numerous plasma cell enrichment techniques have been described. In the paper, we review the cytogenetic approach to identify clinically significant genetic aberrations including the effective use of FISH panels and plasma cell enrichment techniques.

Authors' Contributions

A.D. and M.P. wrote the original draft, S.D. prepared the figures; M.P. and A.D. have full access to all data and the final responsibility for publication. All authors reviewed the manuscript draft submitted for publication.




Publication History

Article published online:
24 April 2023

© 2023. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Thieme Medical and Scientific Publishers Pvt. Ltd.
A-12, 2nd Floor, Sector 2, Noida-201301 UP, India

 
  • References

  • 1 Campo E, Jaffe ES, Cook JR. et al. The International Consensus Classification of Mature Lymphoid Neoplasms: a report from the Clinical Advisory Committee. Blood 2022; 140 (11) 1229-1253
  • 2 Swerdlow SH, Campo E, Pileri SA. et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood 2016; 127 (20) 2375-2390
  • 3 Kumar SK, Rajkumar V, Kyle RA. et al. Multiple myeloma. Nat Rev Dis Primers 2017; 3 (01) 17046
  • 4 Rajan AM, Rajkumar SV. Interpretation of cytogenetic results in multiple myeloma for clinical practice. Blood Cancer J 2015; 5 (10) e365-e365
  • 5 Furukawa Y, Kikuchi J. Molecular pathogenesis of multiple myeloma. Int J Clin Oncol 2015; 20 (03) 413-422
  • 6 Cardona-Benavides IJ, de Ramón C, Gutiérrez NC. Genetic abnormalities in multiple myeloma: prognostic and therapeutic implications. Cells 2021; 10 (02) 336
  • 7 Hanamura I. Multiple myeloma with high-risk cytogenetics and its treatment approach. Int J Hematol 2022; 115 (06) 762-777
  • 8 Rajkumar SV. Multiple myeloma: 2020 update on diagnosis, risk-stratification and management. Am J Hematol 2020; 95 (05) 548-567
  • 9 D'Agostino M, Cairns DA, Lahuerta JJ. et al. Second revision of the International Staging System (R2-ISS) for overall survival in multiple myeloma: a European Myeloma Network (EMN) Report within the HARMONY Project. J Clin Oncol 2022; 40 (29) 3406-3418
  • 9 Rajkumar SV, Tefferi A. Expanding the journal's circulation to oncologists—a new dimension for Mayo Clinic proceedings. Mayo Clin Proc 2007; 82 (08) 915-916
  • 11 Russell SJ, Rajkumar SV. Multiple myeloma and the road to personalised medicine. Lancet Oncol 2011; 12 (07) 617-619
  • 12 Rajkumar SV, Kohli M. Introduction to the symposium on neoplastic hematology and medical oncology. Mayo Clin Proc 2015; 90 (07) 848-849
  • 13 Treatment-of-Newly-Diagnosed-Myelomav16_Nov19FINAL.pdf. Accessed October 10, 2022. Accessed Feb 1, 2023 at: https://static1.squarespace.com/static/5b44f08ac258b493a25098a3/t/5dd6c13ee9a64d38904eabda/1574355263029/Treatment-of-Newly-Diagnosed-Myelomav16_Nov19FINAL.pdf
  • 14 Baysal M, Demirci U, Umit E. et al. Concepts of double hit and triple hit disease in multiple myeloma, entity and prognostic significance. Sci Rep 2020; 10 (01) 5991
  • 15 Shen M, Yang G, Li X, Geng C, Huang Z, Chen W. At least two high-risk cytogenetic abnormalities indicate the inferior outcomes for newly diagnosed multiple myeloma patients: a real-world study in China. Leuk Lymphoma 2021; 62 (12) 2992-3001
  • 16 Walker BA, Mavrommatis K, Wardell CP. et al. A high-risk, double-hit, group of newly diagnosed myeloma identified by genomic analysis. Leukemia 2019; 33 (01) 159-170
  • 17 Talley PJ, Chantry AD, Buckle CH. Genetics in myeloma: genetic technologies and their application to screening approaches in myeloma. Br Med Bull 2015; 113 (01) 15-30
  • 18 Stevens-Kroef M, Weghuis DO, Croockewit S. et al. High detection rate of clinically relevant genomic abnormalities in plasma cells enriched from patients with multiple myeloma. Genes Chromosomes Cancer 2012; 51 (11) 997-1006
  • 19 Chng WJ, Dispenzieri A, Chim CS. et al; International Myeloma Working Group. IMWG consensus on risk stratification in multiple myeloma. Leukemia 2014; 28 (02) 269-277
  • 20 Anguiano A, Tuchman SA, Acharya C. et al. Gene expression profiles of tumor biology provide a novel approach to prognosis and may guide the selection of therapeutic targets in multiple myeloma. J Clin Oncol 2009; 27 (25) 4197-4203
  • 21 Abós B, Wang T, Castro R. et al. Distinct differentiation programs triggered by IL-6 and LPS in teleost IgM(+) B cells in the absence of germinal centers. Sci Rep 2016; 6 (01) 30004
  • 22 Kishimoto RK, de Freitas SLVV, Ratis CA, Borri D, Sitnik R, Velloso EDRP. Validation of interphase fluorescence in situ hybridization (iFISH) for multiple myeloma using CD138 positive cells. Rev Bras Hematol Hemoter 2016; 38 (02) 113-120
  • 23 López de Frutos L, Álvarez Y, Armengol G, Caballín MR. New mitogens in cultures for multiple myeloma cytogenetic analysis. Leuk Lymphoma 2013; 54 (11) 2548-2550
  • 24 Sonneveld P, Avet-Loiseau H, Lonial S. et al. Treatment of multiple myeloma with high-risk cytogenetics: a consensus of the International Myeloma Working Group. Blood 2016; 127 (24) 2955-2962
  • 25 Fonseca R, Bergsagel PL, Drach J. et al; International Myeloma Working Group. International Myeloma Working Group molecular classification of multiple myeloma: spotlight review. Leukemia 2009; 23 (12) 2210-2221
  • 26 Fonseca R, Barlogie B, Bataille R. et al. Genetics and cytogenetics of multiple myeloma: a workshop report. Cancer Res 2004; 64 (04) 1546-1558
  • 27 Wahed A, Quesada A, Dasgupta A. . Application of flow cytometry in diagnosis of hematological disorders. In: Hematology and Coagulation. Elsevier; 2020:127–141.
  • 28 Hartmann L, Biggerstaff JS, Chapman DB. et al. Detection of genomic abnormalities in multiple myeloma: the application of FISH analysis in combination with various plasma cell enrichment techniques. Am J Clin Pathol 2011; 136 (05) 712-720
  • 29 Khoenkhoen S, Ádori M, Pedersen GK, Karlsson Hedestam GB. Flow cytometry-based protocols for the analysis of human plasma cell differentiation. Front Immunol 2020; 11: 571321
  • 30 Mercer BR, Rayeroux KC. . Detection of chromosome abnormalities using cytoplasmic immunoglobulin staining and FISH in myeloma. In: Campbell LJ, ed. Cancer Cytogenetics. Vol. 730. Methods in Molecular Biology. Humana Press; 2011:159–171. doi:
  • 31 Yang HS, Montella J, Holt L. et al. Cytoplasmic immunoglobulin fluorescence in situ hybridization (cIg FISH) enhances the quantitative detection of chromosome abnormalities in plasma cell neoplasms compared to conventional FISH. Blood 2010; 116 (21) 1198-1198
  • 32 Gole L, Lin A, Chua C, Chng WJ. Modified cIg-FISH protocol for multiple myeloma in routine cytogenetic laboratory practice. Cancer Genet 2014; 207 (1-2): 31-34
  • 33 Ma ESK, Wang CLN, Wong ATC, Choy G, Chan TL. Target fluorescence in-situ hybridization (target FISH) for plasma cell enrichment in myeloma. Mol Cytogenet 2016; 9 (01) 63
  • 34 Yan Y, Qin X, Liu J. et al. Clonal phylogeny and evolution of critical cytogenetic aberrations in multiple myeloma at single-cell level by QM-FISH. Blood Adv 2022; 6 (02) 441-451
  • 35 Ross FM, Avet-Loiseau H, Ameye G. et al; European Myeloma Network. Report from the European Myeloma Network on interphase FISH in multiple myeloma and related disorders. Haematologica 2012; 97 (08) 1272-1277
  • 36 Ciolino AL, Tang ME, Bryant R. Statistical treatment of fluorescence in situ hybridization validation data to generate normal reference ranges using Excel functions. J Mol Diagn 2009; 11 (04) 330-333
  • 37 Kadam Amare PS, Jain H, Nikalje S. et al. Observation on frequency & clinico-pathological significance of various cytogenetic risk groups in multiple myeloma: an experience from India. Indian J Med Res 2016; 144 (04) 536-543
  • 38 Nishida K, Tamura A, Nakazawa N. et al. The Ig heavy chain gene is frequently involved in chromosomal translocations in multiple myeloma and plasma cell leukemia as detected by in situ hybridization. Blood 1997; 90 (02) 526-534
  • 39 Barwick BG, Gupta VA, Vertino PM, Boise LH. Cell of origin and genetic alterations in the pathogenesis of multiple myeloma. Front Immunol 2019; 10: 1121
  • 40 Manier S, Salem KZ, Park J, Landau DA, Getz G, Ghobrial IM. Genomic complexity of multiple myeloma and its clinical implications. Nat Rev Clin Oncol 2017; 14 (02) 100-113
  • 41 Kumar S, Kaufman JL, Gasparetto C. et al. Efficacy of venetoclax as targeted therapy for relapsed/refractory t(11;14) multiple myeloma. Blood 2017; 130 (22) 2401-2409
  • 42 Pawlyn C, Davies FE. Toward personalized treatment in multiple myeloma based on molecular characteristics. Blood 2019; 133 (07) 660-675
  • 43 Cazaubiel T, Leleu X, Perrot A. et al. Primary plasma cell leukemias displaying t(11;14) have specific genomic, transcriptional, and clinical features. Blood 2022; 139 (17) 2666-2672
  • 44 Kalff A, Khong T, Wall M. et al. A rare case of IGH/MYC and IGH/BCL2 double hit primary plasma cell leukemia. Haematologica 2015; 100 (02) e60-e62
  • 45 Kumar S, Fonseca R, Ketterling RP. et al. Trisomies in multiple myeloma: impact on survival in patients with high-risk cytogenetics. Blood 2012; 119 (09) 2100-2105
  • 46 Chretien ML, Corre J, Lauwers-Cances V. et al. Understanding the role of hyperdiploidy in myeloma prognosis: which trisomies really matter?. Blood 2015; 126 (25) 2713-2719
  • 47 Caers J, Garderet L, Kortüm KM. et al. European Myeloma Network recommendations on tools for the diagnosis and monitoring of multiple myeloma: what to use and when. Haematologica 2018; 103 (11) 1772-1784
  • 48 Ye JC, Chen L, Chen J. et al. Aneuploidy is associated with inferior survival in relapsed refractory multiple myeloma patients. Blood 2019; 134 (Supplement_1): 4360-4360
  • 49 Schmidt TM, Barwick BG, Joseph N. et al. Gain of chromosome 1q is associated with early progression in multiple myeloma patients treated with lenalidomide, bortezomib, and dexamethasone. Blood Cancer J 2019; 9 (12) 94
  • 50 Bisht K, Walker B, Kumar SK. et al. Chromosomal 1q21 abnormalities in multiple myeloma: a review of translational, clinical research, and therapeutic strategies. Expert Rev Hematol 2021; 14 (12) 1099-1114
  • 51 D'Agostino M, Ruggeri M, Aquino S. et al. Impact of gain and amplification of 1q in newly diagnosed multiple myeloma patients receiving carfilzomib-based treatment in the forte trial. Blood 2020; 136 (Supplement (Suppl. 01) 38-40
  • 52 Treon SP, Maimonis P, Bua D. et al. Elevated soluble MUC1 levels and decreased anti-MUC1 antibody levels in patients with multiple myeloma. Blood 2000; 96 (09) 3147-3153
  • 53 Sawyer JR, Tricot G, Lukacs JL. et al. Genomic instability in multiple myeloma: evidence for jumping segmental duplications of chromosome arm 1q. Genes Chromosomes Cancer 2005; 42 (01) 95-106
  • 54 Legartova S, Krejci J, Harnicarova A, Hajek R, Kozubek S, Bartova E. Nuclear topography of the 1q21 genomic region and Mcl-1 protein levels associated with pathophysiology of multiple myeloma. Neoplasma 2009; 56 (05) 404-413
  • 55 Hebraud B, Leleu X, Lauwers-Cances V. et al. Deletion of the 1p32 region is a major independent prognostic factor in young patients with myeloma: the IFM experience on 1195 patients. Leukemia 2014; 28 (03) 675-679
  • 56 Avet-Loiseau H, Li C, Magrangeas F. et al. Prognostic significance of copy-number alterations in multiple myeloma. J Clin Oncol 2009; 27 (27) 4585-4590
  • 57 Boyd KD, Ross FM, Walker BA. et al; NCRI Haematology Oncology Studies Group. Mapping of chromosome 1p deletions in myeloma identifies FAM46C at 1p12 and CDKN2C at 1p32.3 as being genes in regions associated with adverse survival. Clin Cancer Res 2011; 17 (24) 7776-7784
  • 58 Corre J, Perrot A, Caillot D. et al. del(17p) without TP53 mutation confers a poor prognosis in intensively treated newly diagnosed patients with multiple myeloma. Blood 2021; 137 (09) 1192-1195
  • 59 Merz M, Hielscher T, Seckinger A. et al. Baseline characteristics, chromosomal alterations, and treatment affecting prognosis of deletion 17p in newly diagnosed myeloma. Am J Hematol 2016; 91 (11) E473-E477
  • 60 Fonseca R, Oken MM, Harrington D. et al. Deletions of chromosome 13 in multiple myeloma identified by interphase FISH usually denote large deletions of the q arm or monosomy. Leukemia 2001; 15 (06) 981-986
  • 61 Weinhold N, Kirn D, Seckinger A. et al. Concomitant gain of 1q21 and MYC translocation define a poor prognostic subgroup of hyperdiploid multiple myeloma. Haematologica 2016; 101 (03) e116-e119
  • 62 Glitza IC, Lu G, Shah R. et al. Chromosome 8q24.1/c-MYC abnormality: a marker for high-risk myeloma. Leuk Lymphoma 2015; 56 (03) 602-607
  • 63 Abdallah N, Baughn LB, Rajkumar SV. et al. Implications of MYC rearrangements in newly diagnosed multiple myeloma. Clin Cancer Res 2020; 26 (24) 6581-6588
  • 64 Caracciolo D, Scionti F, Juli G. et al. Exploiting MYC-induced PARPness to target genomic instability in multiple myeloma. Haematologica 2021; 106 (01) 185-195
  • 65 Kim H, Moon HW, Hur M, Yun YM, Park CM, Lee MH. Variant Burkitt-type translocation (8;22)(q24;q11) in plasma cell myeloma. Korean J Hematol 2011; 46 (02) 135-138
  • 66 Kyle RA, Therneau TM, Rajkumar SV. et al. A long-term study of prognosis in monoclonal gammopathy of undetermined significance. N Engl J Med 2002; 346 (08) 564-569
  • 67 Lakshman A, Paul S, Rajkumar SV. et al. Prognostic significance of interphase FISH in monoclonal gammopathy of undetermined significance. Leukemia 2018; 32 (08) 1811-1815
  • 68 Kyle RA, Remstein ED, Therneau TM. et al. Clinical course and prognosis of smoldering (asymptomatic) multiple myeloma. N Engl J Med 2007; 356 (25) 2582-2590
  • 69 Mateos MV, Kumar S, Dimopoulos MA. et al. International Myeloma Working Group risk stratification model for smoldering multiple myeloma (SMM). Blood Cancer J 2020; 10 (10) 102
  • 70 Rajkumar SV, Gupta V, Fonseca R. et al. Impact of primary molecular cytogenetic abnormalities and risk of progression in smoldering multiple myeloma. Leukemia 2013; 27 (08) 1738-1744
  • 71 Neben K, Jauch A, Hielscher T. et al. Progression in smoldering myeloma is independently determined by the chromosomal abnormalities del(17p), t(4;14), gain 1q, hyperdiploidy, and tumor load. J Clin Oncol 2013; 31 (34) 4325-4332
  • 72 Papadhimitriou SI, Terpos E, Liapis K. et al. The cytogenetic profile of primary and secondary plasma cell leukemia: etiopathogenetic perspectives, prognostic impact and clinical relevance to newly diagnosed multiple myeloma with differential circulating clonal plasma cells. Biomedicines 2022; 10 (02) 209
  • 73 Mina R, D'Agostino M, Cerrato C, Gay F, Palumbo A. Plasma cell leukemia: update on biology and therapy. Leuk Lymphoma 2017; 58 (07) 1538-1547
  • 74 Fernández de Larrea C, Kyle R, Rosiñol L. et al. Primary plasma cell leukemia: consensus definition by the International Myeloma Working Group according to peripheral blood plasma cell percentage. Blood Cancer J 2021; 11 (12) 192
  • 75 Janssen JW, Vaandrager JW, Heuser T. et al. Concurrent activation of a novel putative transforming gene, myeov, and cyclin D1 in a subset of multiple myeloma cell lines with t(11;14)(q13;q32). Blood 2000; 95 (08) 2691-2698