Next Article in Journal
Prenatal Detection of Uniparental Disomies (UPD): Intended and Incidental Finding in the Era of Next Generation Genomics
Next Article in Special Issue
46,XY,r(8)/45,XY,−8 Mosaicism as a Possible Mechanism of the Imprinted Birk-Barel Syndrome: A Case Study
Previous Article in Journal
TaYS1A, a Yellow Stripe-Like Transporter Gene, Is Required for Wheat Resistance to Puccinia striiformis f. sp. Tritici
Previous Article in Special Issue
In Silico Model for Chemical-Induced Chromosomal Damages Elucidates Mode of Action and Irrelevant Positives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Tangle of Genomic Aberrations Drives Multiple Myeloma and Correlates with Clinical Aggressiveness of the Disease: A Comprehensive Review from a Biological Perspective to Clinical Trial Results

by
Mariarosaria Sessa
*,
Francesco Cavazzini
,
Maurizio Cavallari
,
Gian Matteo Rigolin
and
Antonio Cuneo
Hematology Section, Department of Medical Sciences, Azienda Ospedaliero-Universitaria, Arcispedale S.Anna, University of Ferrara, 44121 Ferrara, Italy
*
Author to whom correspondence should be addressed.
These authors jointly supervised this work.
Submission received: 3 November 2020 / Revised: 24 November 2020 / Accepted: 1 December 2020 / Published: 3 December 2020
(This article belongs to the Special Issue Causes and Consequences of Chromosomal Aberrations)

Abstract

:
Multiple myeloma (MM) is a genetically heterogeneous disease, in which the process of tumorigenesis begins and progresses through the appearance and accumulation of a tangle of genomic aberrations. Several are the mechanisms of DNA damage in MM, varying from single nucleotide substitutions to complex genomic events. The timing of appearance of aberrations is well studied due to the natural history of the disease, that usually progress from pre-malignant to malignant phase. Different kinds of aberrations carry different prognostic significance and have been associated with drug resistance in some studies. Certain genetic events are well known to be associated with prognosis and are incorporated in risk evaluation in MM at diagnosis in the revised International Scoring System (R-ISS). The significance of some other aberrations needs to be further explained. Since now, few phase 3 randomized trials included analysis on patient’s outcomes according to genetic risk, and further studies are needed to obtain useful data to stratify the choice of initial and subsequent treatment in MM.

1. Introduction

Multiple myeloma (MM) is a plasma cell disorder, accounting in term of prevalence for about 10% of hematological malignancies. MM is a genetically heterogenous disease arising from the accumulation of genomic events over the years [1]. The model of clonal progression from a pre-malignant stage of monoclonal gammopathy of uncertain significance (MGUS) through an asymptomatic stage named smoldering multiple myeloma (SMM) to MM and plasma cell leukemia (PCL) is well studied [2,3,4]. The accumulation of mutations and chromosome abnormalities over the time [5] favor us in considering MM a model of cancer genesis and evolution.
The process of tumor genesis and progression is triggered by several recurrent events at the genome and chromosome level. Some events typically occur at diagnosis, whereas others could be detected in most patients in late stages of the disease [6]. Furthermore, the concurrence of several and genetically heterogeneous subclones within MM cells—in a context of selection pressure—complicates the interpretation of the genetic of the tumor for intrinsic spatial [7,8] and population heterogeneity [9,10,11]. In recent years, whole-exome (WES) and whole-genome sequencing (WGS) studies highlighted the complex genomic aberrations underlying the pathogenesis of the disease [12,13,14,15].
This review aims to present the tangle of genomic events that drive plasma cell disorders, particularly MM, and to focus on their clinical significance.

2. Landscape of Genomic Damage in MM

2.1. Timing of Genome Aberrations in MM

MM has been studied by systematic analysis of target sequencing, WES, and single nucleotide polymorphisms (SNP) array, to identify DNA aberrations recurring at high and intermediate frequencies, with the aim to appreciate pathogenetic dysregulated pathways and potential therapy targets. The analyses revealed in MM a somatic mutation rate of 1.6 mutations per MB [11,13,14,16,17,18,19,20,21,22].
Plasma cell disorders are genetically heterogeneous diseases and their clonal progression occurs in sequential phases [11]. Hyperdiploid (HD) events are characterized by the acquisition of chromosome trisomy. Frequently, trisomy occurs in chromosome 3, 5, 7, 9, 11, 15, 19, 21 [19,23]. HD is the main event identified in early stage MM [11]. Non-HD events occurring in early stage MM pathogenesis are characterized by the well-known translocations typically affecting the genes encoding immunoglobulin (Ig) heavy chains (IGH), mainly t(4;14), t(6;14), t(11;14), t(14;16), and t(14;20) [19]. HD MM carries a lower prevalence of primary translocations, as opposed to non-HD MM. Due to their different pathogenesis, they are recognized as the two main genetic subtypes of MM. Secondary events, detected generally in later stages, are essential for tumor progression and consist of chromosomal translocations, copy-number variations (CNV) and single-nucleotide somatic variations that affect several signaling pathways, cell-cycle regulators, and DNA-damage repair mechanisms [19].
Aktas Samur recently described the chronology of copy number aberrations in 164 MGUS and 336 newly diagnosed MM. HD MM seems to originate from different events if compared with non-HD MM. In HD MM, CNVs are also clonal at the MGUS stage, ratifying that these are early events leading to transformation of normal plasma cell to pathologic MGUS cell but are not sufficient to generate MM proliferating cells. The proposed model of HD myeloma genesis and progression suggests the initial gain of at least two copy of chromosome 9, 15, and 19 also present in the MGUS stage; some patients then acquires trisomy of chromosome 6, or chromosome 18, or chromosome 21, while other patients lose chromosome 13 and/or gain 1q (about 20% of patients show no additional CNVs). In non-HD MM, IgH translocations are the earliest event, also identified in MGUS, and most patients do not accumulate other additional changes. In both groups, late catastrophic genomic events can occur, leading to complex deletions, irrespective of initial aberrations [24]. A mention must be referred to TP53 aberrations. Both mutations of the TP53 gene and deletions of chromosome 17p13 containing the TP53 gene are late events in MM history [9,25]. Deletion of 17p occurs with a frequency of about 9.5% in newly diagnosed symptomatic MM patients, rising up to 50% in primary plasma cell leukemia and to 75% in secondary plasma cell leukemia patients [19,26]. Similarly, TP53 mutations occur in 5–8% of newly diagnosed symptomatic MM patients, rising up to 25% in PCL [26,27,28,29,30]. TP53 mutations usually appears after or concurrently with deletion 17p13 [31]. Conversely, TP53 mutations are identified in one third of 17p deleted patients [27].
Damages arise in clone or sub-clone in MM. Early mutations in MM evolution are expected to be clonal. Mutations mostly occurring at the sub-clonal level arise later in MM progression. Nevertheless, several driver gene mutations can be found in MM at the sub-clonal level, underlining the heterogeneity of the cell population. For example, Bolli et al. identified sub-clonal mutations of the driver genes KRAS, NRAS, TP53. TP53 was sub-clonal in 26.7% and clonal in 11% of case. No significant differences were identified in prognosis when a mutation was identified at the clonal or sub-clonal level [5]. Data are lacking about clonal selection induced by treatment in MM. An analysis by Corre et al. evaluated 43 homogeneously treated MM patients for mutational profiles at the time of diagnosis and at relaps [32]. No specific or significant events in mutational status were found after treatment neither at relapse. Every patient seems to evolve differently, with chemoresistance and relapse driven by acquired mutations or selection of pre-existing subclones [32].

2.2. Mechanisms of DNA Damage in MM

In MM, genomic aberrations arise from various types of DNA damage (Figure 1). Nucleotide damage (ND) and single strand breaks (SSB) hesitate in single base substitutions. Double strand breaks (DSB) often alter chromosome structure.
Mutational events as kataegis and complex genome events (CGE) were described in MM [11,16,17,18,19,20,21,22]. Kataegis and, in general, “apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like” (APOBEC) family-related signatures (a signature with C > T mutations in regions rich in 5-methyl-CpG and another one consisting C > T and C > G mutations in TCN trinucleotide repeats, which results from overactivation of cytidine deaminase activity) were observed in MM [5,11,19].
All the CGE described in MM, chromothripsis, chromoanasynthesis, and chromoplexy—also grouped together under the name of chromoanagenesis—arise from DSB [33]. SNP-microarray and WGS are able to identify these CGE also when localized genomic aberrations are not detected by conventional cytogenetic and Fluorescent in situ hybridization (FISH) analysis [17,34,35]. Genomics events bring a rapid genome variation and could also play a role in genome adaptive evolution [36].
Alterations of ploidy and translocations are frequent as a result of a disfunction in control mechanisms in late mitosis [37]. HD and translocations are the most common events historically described as driver events in MM.
Table 1 reports most frequent DNA damage events, as described in recent genomic studies.

2.2.1. Single-Nucleotide Variants and Indels

WES identified recurrent somatic mutations in MM that drive pathogenesis. Three studies, including 203, 67, and 463 patients respectively, reported that the most frequently involved genes belong to the MAPK pathway, including KRAS, NRAS, and BRAF, mutated in about 40% of cases [10,11,38]. KRAS, NRAS, and BRAF were clonal in about 70% of the patients and sub-clonal in 30% in a cohort f 203 mixed patients, but treatment can select stronger clones, so additional studies are needed to define early from late events [10]. NF-KB is the second most affected pathway in MM. Mutations in this pathway can be found in about 20% of cases, in genes such as TRAF3, CYL, LTB, IKBKB, BIRC2, BIRC3, CARD11, and TRAF3IP1 [10,38,49]. About 15% of MM patients harbored mutations in genes involved in the DNA-repair pathway (TP53, ATM, and ATR) [38]. Recently, TP53 gene mutations were specifically studied with WES in 54 MM samples, due to the well-known poor prognostic role; 7/54 harbored TP53 mutation, 5/54 TP53 mutation together with del(17p). Among them, only 1 patient had TP53 mutation at diagnosis [39]. In addition, tumor suppressor genes DIS3 and FAM46C are frequently mutated in MM together with genes involved in B-cell lineage differentiation as PRDM1, IRF4, LTB, and SP140 [9,10,38].
Recent studies recognized an association between CRBN expression levels and response to IMIDs in MM. Therefore, SNVs in the CRBN gene have been investigated, but their role remains controversial due to the small number of patients [50,51].

2.2.2. Copy Number Variations

Copy number variations (CNVs) can be primary, such as HD genome, or secondary events in MM, ranging from fragments to chromosomal arm deletions or amplifications [43,52,53].
CNVs can be located in focal regions, with high recurrence and contribution in MM pathogenesis. Recurrent and focal CNVs comprise amplifications and deletions and could be associated with prognostic significance [10,53].
Chromosome level copy number variation frequently occurs in newly diagnosed MM. Most frequent amplifications found with chromosome banding analysis or FISH are gain of 1q (31.3%), 6p (15.2%), and 11q (10.5%) [42] and loss of 16q (28.3%), 1p (24%), 14q (23.4%), 8p (22%), 6q (20.8%), 16p (14.7%), 12p (13%) [42]. Deletion of chromosome 17 p arm is found in about 10% of newly diagnosed MM, and up to 80% in later stages [40,41]. Mutations in TP53 gene coexist in 25–40% of patients with del(17p) [27,38]. The most frequent gains by SNP array analysis reported in a study by Walker were amp1q (36%), trisomy 15 (36.8%), 9 (35.9%), 19 (33.3%), 5 (33.3%), 3 (27.2%), 11 (24.6%), 7 (21%), 21 (12.3%), and X (8.7%); the most frequent deletions were 13q (59%), 14q (39%), 16q (35%), 6q (33%), 1p (30%), 8p (25%), 22 (18%), 12p (15%), 20 (12%), and 17p (7%) [43]. A recent analysis confirmed that del13 is the most frequent CNV (60%), followed by 1q gain (37%) and 14q deletion (32%). Deletion of chromosome 13 and 11 gain are early events in MM history [24].
HD is defined as a number of chromosomes ranging from 48 and 74. HD is a primary event in clonal progression and is identified in about 50% of cases of MGUS, SMM, and MM at diagnosis [19,24,44,45,46]. As a hypothesis, the gain of such a wide quantity of genetic material could be the result of a single catastrophic mitotic event [37]. In HD MM patients, the most frequent events are gains in chromosome 19 (95%), 15 (90%), and 9 (90%). More than 96% of patients carry gains of at least two of these three chromosomes. The other odd number chromosomes (5, 11, 3, 7, 21) follow in frequency. Moreover, Del13 is detected in 37% of HD MM patients [24]. HD cells can sometimes harbor coexisting MYC translocation [19,54].

2.2.3. Chromosomal Translocations

Chromosomal translocations can occur early in MM pathogenesis, also being detected in the MGUS stage [55,56,57]. Translocations, frequently occurring during class-switch recombination, mostly affect the IGH locus 14q32.33 on chromosome 14, and result in upregulation of several oncogenes under control of IGH enhancer [58,59]. The most frequently recurring translocation is t(11;14), with 15–20% of prevalence. t(4;14), t(14;16), t(6;14), and t(14;20) occur in 15%, 5%, 1–2%, and 1% of multiple myeloma patients, respectively [19]. Partner genes can be mutated in 10–25% of cases, probably as a result of somatic hypermutation events [19].
Apart from these main translocations, chromosomal translocations involving MYC (and leading to its overexpression) can be found as secondary genomic events in 15–20% of patients with newly diagnosed MM, but only in 3–4% of patients with MGUS or SMM [19,47,48]. Translocations of MYC are identified in about 65% of MM with HD pattern MM [38]. The main partners of MYC translocations are the IGH genes (16.5%), IGL (16.5%), and IGK (6%), but also FAM46C (9.5%), FOXO3 (6%), and BMP6 (3.5%) [19]. These translocations are associated with the mechanism of kataegis [60].

2.2.4. Mutation Clusters

Kataegis is a rare genome event occurring as a consequence of deregulated APOBEC activity on single-stranded DNA (ssDNA), leading to localized hypermutation around the breakpoint sites, before their reparation [13,61]. Cluster of hypermutation are usually several hundred base pair long [13,60,61]. The APOBEC family comprises cytidine-deaminase enzymes targeting ssDNA with high enrichment of C > G and C > T mutations during DNA replication (APOBEC signature), resulting in replicative asymmetry [60,62].
Bolli et al. first identified clusters of kataegis in MM samples [11]. In whole exome sequencing (WES) of 463 MM patients, kataegis was identified in sites of MYC translocation and in other sites. Furthermore, Walker and colleagues observed different complex signatures also associated with APOBEC family activity and eventually linked to deregulation of MAF and MAFB expression. APOBEC signatures were related to poor prognosis in patients enrolled in UK Myeloma XI study [19].

2.2.5. Complex Genome Events

Chromothripsis is a CGE consisting in a major genome aberration, defined by the presence of massive rearrangements, clustered in localized chromosomal regions. Ten to hundreds of rearrangements accumulate in a rapid process generated by a single catastrophic event [63,64,65,66]. The classical hallmark of chromothripsis is the oscillation between two copy-number states [67]. Chromothripsis arises from two main mechanisms: the encapsulation of chromosomes in aberrant structures called micronuclei, with subsequent fragmentation and reassembly in a single chromatid, [65] and the fragmentation of DNA bridges formed between dicentric chromosomes during telomere crisis [68,69]. The PCAWG Consortium analyzed whole-genome sequences (WGS) of 2658 tumor samples including 38 cancer types searching for chromothripsis. The analysis revealed that chromothripsis is very common in cancer with a frequency greater than expected from previous studies, probably due to the presence of frequent additional aberrations which can mask the classical pattern [63]. In large MM patient sets, chromothripsis was detected at diagnosis with high-resolution single nucleotide polymorphism (SNP) arrays with a prevalence between 1.3% [20] and 10.0% [18] and was linked to poor prognosis in terms of progression free survival and overall survival [15,18,20]. In a recent low coverage WGS research, chromothripsis was identified in 24% of 752 newly diagnosed MM patients [15]. Chromosomes most frequently involved in MM were chromosome 1, 11, 12 [21], and 17 [18] even if other chromosomes were occasionally involved [16,17,18]. A recent analysis of WGS of MM patients revealed that chromothripsis can be a driver event in MM pathogenesis and mostly occur early in the history of the disease [17]. Furthermore, chromothripsis detection could also be important at relapse, as a next generations sequencing analysis showed a high prevalence of chromothripsis in resistant MM cell lines and in MM patients, and it was also correlated with resistance to bortezomib [21].
Chromoanasynthesis is another CGE arising from a single-step event during defective DNA replication. The block of template switching at the replicative fork site leads to a regional copy-number gain and to the insertion of short nucleotide sequences at the breakpoint junctions [36,70,71,72]. The underlining mechanism of incorporating fragments of DNA can be the same as in chromothripsis with defective DNA replication occurring in micronuclei. [73]. Chromoanasynthesis originates from errors in two DNA-repair mechanisms, fork stalling plus template switching, and microhomology-mediated break-induced replication, during endogenous or exogenous stress conditions [71,72,74]. Very few studies report chromoanasynthesis data for MM. Although it can be supposed to be a significant event in disease pathogenesis and progression, at this time, no conclusions can be done [16,22]. Recently, templated insertions were identified as the second most frequent complex event in 19% of 752 newly diagnosed MM patients [15].
Chromoplexy is a large aberration, first described in the prostate cancer genome, arising from a single catastrophic event [75,76]. It is defined by the presence of close chains of translocations involving many chromosomes (up to eight), with little or no copy number aberration. Chromoplexy raises from DNA double strand breaks that pair each other at breakpoint ends in a sequence of inter- and intra-chromosomal translocations [77,78]. The process could be triggered by physical clustering of breakpoint sites during DNA replication as in the micronuclei model [36,79]. In MM, chromoplexy was identified with WGS if 5 of 107 newly diagnosed patients, and involved more than 6 chromosomes [18]. A longitudinal WGS study reported chromoplexy in 3 of 67 patients, as a late event in MM history, with one patient showing at progression from asymptomatic to symptomatic MM and two patients at relapse [17]. The last low coverage WGS research reported chromoplexy in 11% of 752 newly diagnosed MM patients, confirming previous data [15].

3. Clinical Implications of Genome Aberrations in MM

Genetic events in MM can carry specific clinical significance, in terms of drug targeting, drug resistance, and prognosis.

3.1. Impact on Prognosis of Genome Aberrations and Clinical Trials Results

The most advanced results in stratifying MM patients are based on genomic lesions, derived by the Myeloma XI trial. A new prognostic model was defined, adding genomic events of relevant prognostic significance to the algorithm of the International Staging System (ISS). This score then additionally combined traditional ISS and LDH levels in a new score called revised-ISS, nowadays commonly used to stratify prognosis in newly diagnosed MM patients [38,80,81]. At diagnosis, a high risk disease based on genomic aberrations is defined by the presence of at least one between del(17p), t(4;14), t(14;16), with a median OS of 24.5 months. The absence, in a low ISS, defines standard risk with a median OS of 50.5 months [80]. Some international trials also define a category of patients with ultra-high risk basing on karyotype, who harbor more than one high risk lesion [82,83]. Table 2 defines risk stratifications of MM based on karyotype abnormalities used in most recent clinical trials.
Afterwards, other attempts to define prognostic scores were published. Perrot et al. proposed a cytogenetic prognostic score which divides patients in 3 categories based on 6 abnormalities—t(4;14), del(17p), trisomy 5, trisomy 21, 1q gain, del1p32 [84]. Interestingly, the first in literature, a recent WGS and WES study identified a subgroup of MM patients with good outcomes, independently by traditional clinical risk factor (R-ISS, treatment response and MRD negativity). It includes about 17% of newly diagnosed MM patients with lower mutational burden, few deletion events, and low genomic instability [85]. However, to date, these new prognostic tools are not widely applied in clinical trials. Table 3 and Table 4 present outcomes from diagnosis by genomic lesions in transplant eligible and ineligible MM patients, respectively, as reported in most recent studies. Overall, therapy intensification and continuous treatment seems to give significant benefit in high risk patients and in most of the genome aberrations. It is of particular interest that pembrolizumab did not show significant benefits in MM with complex genome aberrations, in contrast with results in other cancers [86,87]. Of note, t(11;14) has been identified as the first predictive marker of drug response to BCL2 inhibitors in MM, because of its association with high expression of BCL2 protein [88].
Table 5 shows a proposal of treatment recommendations based on cytogenetic risk that can be drawn out from randomized clinical trials.

3.2. Drug-Resistance

MM is currently treated with a high number of drugs with distinctive mechanisms of action, used in combinations to optimize treatment and prognosis. The choice depends on age, comorbidities, and prognosis.
However, MM often shows occurrences of relapse and refractoriness, mostly due to drug resistance. Genetic instability and heterogeneity in MM are a main cause of drug resistance. Many studies evaluated the mechanisms of drug resistance in MM. Aberrations such as t(4;14), t(14;16), t(14;20), translocations involving c-MYC, 17p, and 13p deletions, have been associated with inadequate response to current treatments [89]. Translocation (4;14) results in overexpression of FGFR3 and MMSET carrying poor prognosis and resistance to alkylating agents but has been associated with survival benefit in patients treated with the proteasome inhibitor bortezomib [80,90]. Translocation (14;16) and (14;20) takes to overexpression of the MAF genes, leading to adverse clinical outcomes and drug resistance in some studies [91,92]. In patients with translocation t(4;14), the overexpression of FGFR3 takes to resistance to dexamethasone, but this is not the only one mechanism of drug resistance in MM cells carrying this abnormality [91,92]. Deletion 17p is associated with advanced stages disease, poor prognosis, and treatment failure due to dysregulation of TP53 ability to control cell cycle and apoptosis in response to DNA damage, as the same as TP53 gene mutations [93,94,95].
The progressive improvement of knowledge in MM biology will allow us to design specific drug combinations to overcome drug resistance. In last years, the use of new agents with specific targets is the best response to this therapeutic need.

3.3. Drug Targeting

Within the high number of mutated genes in MM, some of them could be potentially targeted by drugs. But this approach is limited by the presence of subclones harboring different gene mutations that could be paradoxically activated [10]. In order to overcome the effect of clonal heterogeneity and clonal selection, several new immunotherapeutic drugs have been developed, such as autoantibodies (anti-CD38, anti-CD319), chimeric antigen receptor (CAR-T) cells (against B-cell maturation antigen and Immunoglobulin K light chain), and immune-check point inhibitors, but their role in clonal progression needs to be further indagated [96,97,98,99,100,101].
CAR-T cell therapy is nowadays the most promising treatment against MM, under investigation in several clinical trials with preliminary controversial outcomes. Most outstanding results come from trials with CAR-T cells targeting BCMA [97]. Their extensive use is limited by some challenging side effects, i.e., cytokine release syndrome (CRS) and immune effector neurotoxicity syndrome (ICANS), and by their time consuming and expensive production and transportation requirements [102]. However, CAR-T cell therapy results in MM are overall promising, and this strategy needs to be improved.

4. Conclusions

We presented the tangle of genomic aberrations driving MM pathogenesis and progression, and we focused on their clinical impact. The last decade brings significant advantages in the biological knowledge of MM. Cheap and widely applicable sequencing technologies allow us to go through myeloma biology, and most recent clinical trials are taking advantages from preclinical data, building prognostic models that are also based on genome aberrations. To date, a divide between the enormous bulk of available preclinical data and the scalability of these data in the clinical setting still exists.
A better, genomic-based, prognostic model could have the capability to ameliorate patients’ outcomes. A major effort in incorporating a biology-driven approach in clinical trials is warranted. Genomic aberrations could serve to address precise therapy intervention other than for prognostication purposes. The description of clinical effect of genomic aberrations—at the clonal and sub-clonal level—and their impact on therapy response and clinical outcomes could help us in defining precise subset of patients to address with a certain treatment both at diagnosis and relapse, with the intent to create a personalized but evidence-based approach.

Author Contributions

Conceptualization, M.S.; methodology, M.S., G.M.R.; validation, M.S., F.C., M.C., G.M.R., A.C.; formal analysis, M.S., G.M.R, A.C.; software, M.S.; investigation, NA; resources, A.C.; data curation, M.S.; writing—original draft preparation, M.S., F.C., M.C., G.M.R., A.C.; writing—review and editing, all the authors; visualization, all the authors; supervision, G.M.R., A.C.; project administration, M.S.; funding acquisition, NA. NA: not applicable. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

Authors declare no conflict of interest.

References

  1. Rajkumar, S.V. Multiple myeloma: 2020 update on diagnosis, risk-stratification and management. Am. J. Hematol. 2020, 95, 548–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Kyle, R.A.; Larson, D.R.; Therneau, T.M.; Dispensieri, A.; Kumar, S.; Cerhan, J.R.; Rajkumar, S.V. Long-Term Follow-up of Monoclonal Gammopathy of Undetermined Significance. N. Engl. J. Med. 2018, 378, 241–249. [Google Scholar] [CrossRef] [PubMed]
  3. Kyle, R.A.; Remstein, E.D.; Therneau, T.M.; Dispensieri, A.; Kurtin, P.J.; Hodnefield, J.M.; Larson, D.R.; Plevak, M.F.; Jelinek, D.F.; Fonseca, R.; et al. Clinical Course and Prognosis of Smoldering (Asymptomatic) Multiple Myeloma. N. Engl. J. Med. 2007, 356, 2582–2589. [Google Scholar] [CrossRef] [PubMed]
  4. Van Nieuwenhuijzen, N.; Spaan, I.; Raymakers, R.; Peperzak, V. From MGUS to multiple myeloma, a paradigm for clonal evolution of premalignant cells. Cancer Res. 2018, 78, 2449–2456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Bolli, N.; Biancon, G.; Moarii, M.; Gimondi, S.; Li, Y.; de Philippis, C.; Maura, F.; Sathiaseelan, V.; Tai, Y.T.; Mudie, L.; et al. Analysis of the genomic landscape of multiple myeloma highlights novel prognostic markers and disease subgroups. Leukemia 2018, 32, 2604–2616. [Google Scholar] [CrossRef] [PubMed]
  6. Morgan, G.J.; Walker, B.A.; Davies, F.E. The genetic architecture of multiple myeloma. Nat. Rev. Cancer 2012, 12, 335–348. [Google Scholar] [CrossRef]
  7. Coffey, D.G.; Wu, Q.V.; Towlerton, A.M.H.; Ornelas, S.; Morales, A.J.; Xu, Y.; Green, D.J.; Warren, E.H. Ultradeep, targeted sequencing reveals distinct mutations in blood compared to matched bone marrow among patients with multiple myeloma. Blood Cancer J. 2019, 9, 1–4. [Google Scholar] [CrossRef]
  8. Rasche, L.; Chavan, S.S.; Stephens, O.W.; Patel, P.H.; Tytarenko, R.; Ashby, C.; Bauer, M.; Stein, C.; Deshpande, S.; Wardell, C.; et al. Spatial genomic heterogeneity in multiple myeloma revealed by multi-region sequencing. Nat. Commun. 2017, 8. [Google Scholar] [CrossRef]
  9. Manier, S.; Salem, K.Z.; Park, J.; Landau, D.A.; Getz, G.; Ghobrial, I.M. Genomic complexity of multiple myeloma and its clinical implications. Nat. Rev. Clin. Oncol. 2017, 14, 100–113. [Google Scholar] [CrossRef]
  10. Lohr, J.G.; Stojanov, P.; Carter, S.L.; Cruz-Cordillo, P.; Lawrence, M.S.; Auclair, D.; Sougnez, C.; Knoechel, B.; Gould, J.; Saksena, G.; et al. Widespread genetic heterogeneity in multiple myeloma: Implications for targeted therapy. Cancer Cell 2014, 25, 91–101. [Google Scholar] [CrossRef] [Green Version]
  11. Bolli, N.; Avet-Loiseau, H.; Wedge, D.C.; Van Loo, P.; Alexandrov, L.B.; Martincorena, I.; Dawson, K.J.; Iorio, F.; Nik-Zainal, S.; Bignell, G.R.; et al. Heterogeneity of genomic evolution and mutational profiles in multiple myeloma. Nat. Commun. 2014, 5, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Campbell, P.J.; Getz, G.; Korbel, J.O.; Stuart, J.M.; Jennings, J.L.; Stein, L.D.; Perry, M.D.; Nahal-Bose, H.K.; Ouelette, B.F.F.; Li, C.H.; et al. Pan-cancer analysis of whole genomes. Nature 2020, 578, 82–93. [Google Scholar] [CrossRef] [Green Version]
  13. Lawrence, M.S.; Stojanov, P.; Polak, P.; Kryukov, G.V.; Cibulskis, K.; Sivachenko, A.; Carter, S.L.; Stewart, C.; Mermel, C.H.; Roberts, S.A.; et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 2013, 499, 214–218. [Google Scholar] [CrossRef] [PubMed]
  14. Lawrence, M.S.; Stojanov, P.; Mermel, C.H.; Robinson, J.T.; Garraway, L.A.; Golub, T.R.; Meyerson, M.; Gabriel, S.B.; Lander, E.S.; Getz, G. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 2014, 505, 495–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Rustad, E.H.; Yellapantula, V.D.; Glodzik, D.; Maclachlan, K.H.; Diamond, B.; Boyle, E.M.; Ashby, C.; Blaney, P.; Gundem, G.; Hultcrantz, M.; et al. Revealing the Impact of Structural Variants in Multiple Myeloma. Blood Cancer Discov. 2020, 1, 258–273. [Google Scholar] [CrossRef]
  16. Berry, N.K.; Dixon-McIver, A.; Scott, R.J.; Rowlings, P.; Enjeti, A.K. Detection of complex genomic signatures associated with risk in plasma cell disorders. Cancer Genet. 2017, 218–219, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Maura, F.; Bolli, N.; Angelopoulos, N.; Dawson, K.J.; Leongamornlert, D.; Martincorena, I.; Mitchell, T.J.; Fullam, A.; Gonzales, S.; Szalat, R.; et al. Genomic landscape and chronological reconstruction of driver events in multiple myeloma. Nat. Commun. 2019, 10. [Google Scholar] [CrossRef] [Green Version]
  18. Kaur, G.; Gupta, R.; Mathur, N.; Rani, L.; Kumar, L.; Sharma, A.; Singh, V.; Gupta, A.; Sharma, O.D. Clinical impact of chromothriptic complex chromosomal rearrangements in newly diagnosed multiple myeloma. Leuk. Res. 2019, 76, 58–64. [Google Scholar] [CrossRef]
  19. Walker, B.A.; Wardell, C.P.; Murison, A.; Boyle, E.M.; Begum, D.B.; Dahir, N.M.; Proszek, P.Z.; Melchor, L.; Pawlyn, C.; Kaiser, M.F.; et al. APOBEC family mutational signatures are associated with poor prognosis translocations in multiple myeloma. Nat. Commun. 2015, 6. [Google Scholar] [CrossRef]
  20. Magrangeas, F.; Avet-Loiseau, H.; Munshi, N.C.; Minvielle, S. Chromothripsis identifies a rare and aggressive entity among newly diagnosed multiple myeloma patients. Blood 2011, 118, 675–678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Lee, K.J.; Lee, K.H.; Yoon, K.A.; Sohn, J.Y.; Lee, E.; Lee, H.; Eom, H.S.; Kong, S.Y. Chromothripsis in Treatment Resistance in Multiple Myeloma. Genomics Inform. 2017, 15, 87–97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Saxe, D.; Seo, E.J.; Beaulieu Bergeron, M.; Han, J.Y. Recent advances in cytogenetic characterization of multiple myeloma. Int. J. Lab. Hematol. 2019, 41, 5–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Chng, W.J.; Kumar, S.; VanWier, S.; Ahmann, G.; Price-Troska, T.; Henderson, K.; Chung, T.H.; Kim, S.; Mullingan, G.; Bryant, B.; et al. Molecular dissection of hyperdiploid multiple myeloma by gene expression profiling. Cancer Res. 2007, 67, 2982–2989. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Aktas Samur, A.; Minvielle, S.; Shammas, M.; Fulciniti, M.; Magrangeas, F.; Richardson, P.G.; Moreau, P.; Attal, M.; Anderson, K.C.; Parmigiani, G.; et al. Deciphering the chronology of copy number alterations in Multiple Myeloma. Blood Cancer J. 2019, 9, 1–10. [Google Scholar] [CrossRef] [PubMed]
  25. Jovanović, K.K.; Escure, G.; Demonchy, J.; Willaume, A.; Van de Wyngaert, Z.; Farhat, M.; Chauvet, P.; Facon, T.; Quesnel, B.; Manier, S. Deregulation and targeting of TP53 pathway in multiple myeloma. Front. Oncol. 2019, 9. [Google Scholar] [CrossRef] [PubMed]
  26. Chesi, M.; Robbiani, D.F.; Sebag, M.; Chng, W.J.; Affer, M.; Tiedemann, R.; Valdez, R.; Palmer, S.E.; Haas, S.S.; Stewart, A.K.; et al. AID-Dependent Activation of a MYC Transgene Induces Multiple Myeloma in a Conditional Mouse Model of Post-Germinal Center Malignancies. Cancer Cell 2008, 13, 167–180. [Google Scholar] [CrossRef] [Green Version]
  27. Lodé, L.; Eveillard, M.; Trichet, V.; Soussi, T.; Wuillème, S.; Richebourg, S.; Magrangeas, F.; Ifrah, N.; Campion, L.; Traullé, C.; et al. Mutations in TP53 are exclusively associated with del(17p) in multiple myeloma. Haematologica 2010, 95, 1973–1976. [Google Scholar] [CrossRef] [Green Version]
  28. Boyd, K.D.; Ross, F.M.; Tapper, W.J.; Chiecchio, L.; Dagrada, G.; Konn, Z.J.; Gonzales, D.; Walker, B.A.; Hockley, S.L.; Wardell, C.P.; et al. The clinical impact and molecular biology of del(17p) in multiple myeloma treated with conventional or thalidomide-based therapy. Genes Chromosom. Cancer 2011, 50, 765–774. [Google Scholar] [CrossRef]
  29. Walker, B.A.; Mavrommatis, K.; Wardell, C.P.; Ashby, T.C.; Bauer, M.; Davies, F.E.; Rosenthal, A.; Wang, H.; Qu, P.; Hoering, A.; et al. Identification of novel mutational drivers reveals oncogene dependencies in multiple myeloma. Blood 2018, 132, 587–597. [Google Scholar] [CrossRef]
  30. Lionetti, M.; Barbieri, M.; Manzoni, M.; Fabris, S.; Bandini, C.; Todoerti, K.; Nozza, F.; Rossi, D.; Musto, P.; Bandini, L.; et al. Molecular spectrum of TP53 mutations in plasma cell dyscrasias by next generation sequencing: An italian cohort study and overview of the literature. Oncotarget 2016, 7, 21353–21361. [Google Scholar] [CrossRef] [Green Version]
  31. Chin, M.; Sive, J.I.; Allen, C.; Roddie, C.; Chavda, S.J.; Smith, D.; Blombery, P.; Jones, K.; Ryland, G.L.; Popat, R.; et al. Prevalence and timing of TP53 mutations in del(17p) myeloma and effect on survival. Blood Cancer J. 2017, 7, e610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Corre, J.; Cleynen, A.; Robiou du Pont, S.; Buisson, L.; Bolli, N.; Attal, M.; Munshi, N.; Avet-Loiseau, H. Multiple myeloma clonal evolution in homogeneously treated patients. Leukemia 2018, 32, 2636–2647. [Google Scholar] [CrossRef] [PubMed]
  33. Poot, M. Genes, proteins, and biological pathways preventing chromothripsis. Methods Mol. Biol. 2018, 1769, 231–251. [Google Scholar] [CrossRef] [PubMed]
  34. Berry, N.K.; Scott, R.J.; Rowlings, P.; Enjeti, A.K. Clinical use of SNP-microarrays for the detection of genome-wide changes in haematological malignancies. Crit. Rev. Oncol. Hematol. 2019, 142, 58–67. [Google Scholar] [CrossRef]
  35. Sawyer, J.R. The prognostic significance of cytogenetics and molecular profiling in multiple myeloma. Cancer Genet. 2011, 204, 3–12. [Google Scholar] [CrossRef]
  36. Pellestor, F.; Gatinois, V. Chromoanagenesis: A piece of the macroevolution scenario. Mol. Cytogenet. 2020, 13, 1–9. [Google Scholar] [CrossRef]
  37. Onodera, N.; McCabe, N.R.; Rubin, C.M. Formation of a hyperdiploid karyotype in childhood acute lymphoblastic leukemia. Blood 1992, 80, 203–208. [Google Scholar] [CrossRef] [Green Version]
  38. Walker, B.A.; Boyle, E.M.; Wardell, C.P.; Murison, A.; Begum, D.B.; Dahir, N.B.; Proszek, P.Z.; Johnson, D.C.; Kaiser, M.F.; Melchor, L.; et al. Mutational spectrum, copy number changes, and outcome: Results of a sequencing study of patients with newly diagnosed myeloma. J. Clin. Oncol. 2015, 33, 3911–3920. [Google Scholar] [CrossRef]
  39. Petrackova, A.; Minarik, J.; Sedlarikova, L.; Libigerova, T.; Hamplova, A.; Krhovska, P.; Balcarkova, J.; Pika, T.; Papajik, T.; Kriegova, E. Diagnostic deep-targeted next-generation sequencing assessment of TP53 gene mutations in multiple myeloma from the whole bone marrow. Br. J. Haematol. 2020, 189, e122–e125. [Google Scholar] [CrossRef] [Green Version]
  40. Fonseca, R.; Blood, E.; Rue, M.; Harrington, D.; Oken, M.M.; Kyle, R.A.; Dewald, G.W.; Van Ness, B.; Van Wier, S.A.; Henderson, K.J.; et al. Clinical and biologic implications of recurrent genomic aberrations in myeloma. Blood 2003, 101, 4569–4575. [Google Scholar] [CrossRef] [Green Version]
  41. Tiedemann, R.E.; Gonzalez-Paz, N.; Kyle, R.A.; Santana-Davila, R.; Price-Troska, T.; Van Wier, S.A.; Chng, W.J.; Ketterling, R.P.; Gertz, M.A.; Henderson, K.; et al. Genetic aberrations and survival in plasma cell leukemia. Leukemia 2008, 22, 1044–1052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Avet-Loiseau, H.; Li, C.; Magrangeas, F.; Gourand, W.; Charbonnel, C.; Harousseau, J.L.; Attal, M.; Marit, G.; Mathiot, C.; Facon, T.; et al. Prognostic significance of copy-number alterations in multiple myeloma. J. Clin. Oncol. 2009, 27, 4585–4590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Walker, B.A.; Leone, P.E.; Chiecchio, L.; Dickens, N.J.; Jenner, M.W.; Boyd, K.D.; Johnson, D.C.; Gonzalez, D.; Dagrada, G.P.; Protheroe, R.K.M.; et al. A compendium of myeloma-associated chromosomal copy number abnormalities and their prognostic value. Blood 2010, 116. [Google Scholar] [CrossRef] [PubMed]
  44. Smadja, N.V.; Fruchart, C.; Isnard, F.; Louvet, C.; Dutel, J.L.; Cheron, N.; Grange, M.J.; Monconduit, M.; Bastard, C. Chromosomal analysis in multiple myeloma: Cytogenetic evidence of two different diseases. Leukemia 1998, 12, 960–969. [Google Scholar] [CrossRef] [Green Version]
  45. Debes-Marun, C.S.; Dewald, G.W.; Bryant, S.; Picken, E.; Santana-Dávila, R.; González-Paz, N.; Winkler, J.M.; Kyle, R.A.; Gertz, M.A.; Witzig, T.E.; et al. Chromosome abnormalities clustering and its implications for pathogenesis and prognosis in myeloma. Leukemia 2003, 17, 427–436. [Google Scholar] [CrossRef] [Green Version]
  46. Chng, W.J.; Van Wier, S.A.; Ahmann, G.J.; Winkler, J.M.; Jalal, S.M.; Bergsagel, P.L.; Chesi, M.; Trendle, M.C.; Oken, M.M.; Blood, E.; et al. A validated FISH trisomy index demonstrates the hyperdiploid and nonhyperdiploid dichotomy in MGUS. Blood 2005, 106, 2156–2161. [Google Scholar] [CrossRef] [Green Version]
  47. Avet-Loiseau, H.; Gerson, F.; Magrangeas, F.; Minvielle, S.; Harousseau, J.L.; Bataille, R. Intergroupe Francophone du Myélome. Rearrangements of the c-myc oncogene are present in 15% of primary human multiple myeloma tumors. Blood 2001, 98, 3082–3086. [Google Scholar] [CrossRef] [Green Version]
  48. Walker, B.A.; Wardell, C.P.; Brioli, A.; Boyle, E.; Kaiser, M.F.; Begum, D.B.; Dahir, N.B.; Johnson, D.C.; Ross, F.M.; Davies, F.E.; et al. Translocations at 8q24 juxtapose MYC with genes that harbor superenhancers resulting in overexpression and poor prognosis in myeloma patients. Blood Cancer J. 2014, 4, e191. [Google Scholar] [CrossRef]
  49. Keats, J.J.; Fonseca, R.; Chesi, M.; Schop, R.; Baker, A.; Chng, W.J.; Van Wier, S.; Tiedemann, R.; Shi, C.X.; Sebag, M.; et al. Promiscuous Mutations Activate the Noncanonical NF-κB Pathway in Multiple Myeloma. Cancer Cell 2007, 12, 131–144. [Google Scholar] [CrossRef] [Green Version]
  50. Butrym, A.; Łacina, P.; Rybka, J.; Chaszczewska-Markowska, M.; Mazur, G.; Bogunia-Kubik, K. Cereblon and IRF4 Variants Affect Risk and Response to Treatment in Multiple Myeloma. Arch. Immunol. Ther. Exp. (Warsz) 2016, 64, 151–156. [Google Scholar] [CrossRef] [Green Version]
  51. Huang, P.A.; Beedie, S.L.; Chau, C.H.; Venzon, D.J.; Gere, S.; Kazandjian, D.; Korde, N.; Mailankody, S.; Landgren, O.; Figg, W.D. Cereblon gene variants and clinical outcome in multiple myeloma patients treated with lenalidomide. Sci. Rep. 2019, 9, 1–7. [Google Scholar] [CrossRef] [PubMed]
  52. Walker, B.A.; Leone, P.E.; Jenner, M.W.; Li, C.; Gonzalez, D.; Johnson, D.C.; Ross, F.M.; Davies, F.E.; Morgan, G.J. Integration of global SNP-based mapping and expression arrays reveals key regions, mechanisms, and genes important in the pathogenesis of multiple myeloma. Blood 2006, 108, 1733–1743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Carrasco, D.R.; Tonon, G.; Huang, Y.; Zhang, Y.; Sinha, R.; Feng, B.; Stewart, J.P.; Zhan, F.; Khatry, D.; Protopopova, M.; et al. High-resolution genomic profiles define distinct clinico-pathogenetic subgroups of multiple myeloma patients. Cancer Cell 2006, 9, 313–325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Fonseca, R.; Debes-Marun, C.S.; Picken, E.B.; Dewald, G.W.; Bryant, S.C.; Winkler, J.M.; Blood, E.; Oken, M.M.; Santana-Dávila, R.; González-Paz, N.; et al. The recurrent IgH translocations are highly associated with nonhyperdiploid variant multiple myeloma. Blood 2003, 102, 2562–2567. [Google Scholar] [CrossRef]
  55. Kaufmann, H.; Ackermann, J.; Baldia, C.; Nösslinger, T.; Wieser, R.; Seidl, S.; Sagaster, V.; Gisslinger, H.; Jäger, U.; Pfeilstöcker, M.; et al. Both IGH translocations and chromosome 13q deletions are early events in monoclonal gammopathy of undetermined significance and do not evolve during transition to multiple myeloma. Leukemia 2004, 18, 1879–1882. [Google Scholar] [CrossRef] [Green Version]
  56. Fonseca, R.; Bailey, R.J.; Ahmann, G.J.; Rajkumar, S.V.; Hoyer, J.D.; Lust, J.A.; Kyle, R.A.; Gertz, M.A.; Greipp, P.R.; Dewald, G.W. Genomic abnormalities in monoclonal gammopathy of undetermined significance. Blood 2002, 100, 1417–1424. [Google Scholar] [CrossRef]
  57. Avet-Loiseau, H.; Facon, T.; Daviet, A.; Godon, C.; Rapp, M.J.; Harousseau, J.L.; Grosbois, B.; Bataille, R. 14q32 Translocations and Monosomy 13 Observed in Monoclonal Gammopathy of Undetermined Significance Delineate a Multistep Process for the Oncogenesis of Multiple Myeloma 1. CANCER Res. 1999, 59, 4546–4550. [Google Scholar]
  58. González, D.; Van Der Burg, M.; García-Sanz, R.; Fenton, J.A.; Langerak, A.W.; González, M.; van Dongen, J.J.M.; San Miguel, J.F.; Morgan, G.J. Immunoglobulin gene rearrangements and the pathogenesis of multiple myeloma. Blood 2007, 110, 3112–3121. [Google Scholar] [CrossRef]
  59. Bergsagel, P.L.; Kuehl, W.M.; Zhan, F.; Sawyer, J.; Barlogie, B.; Shaughnessy, J., Jr. Cyclin D dysregulation: An early and unifying pathogenic event in multiple myeloma. Blood 2005, 106, 296–303. [Google Scholar] [CrossRef] [Green Version]
  60. Haradhvala, N.J.; Polak, P.; Stojanov, P.; Covington, K.R.; Shinbrot, E.; Hess, J.M.; Rheinbay, E.; Kim, J.; Maruvka, Y.E.; Braunstein, L.Z.; et al. Mutational Strand Asymmetries in Cancer Genomes Reveal Mechanisms of DNA Damage and Repair. Cell 2016, 164, 538–549. [Google Scholar] [CrossRef] [Green Version]
  61. Alexandrov, L.B.; Nik-Zainal, S.; Wedge, D.C.; Aparicio, S.A.J.R.; Behjati, S.; Biankin, A.V.; Bignell, G.R.; 1 Bolli, N.; Borg, A.; Børresen-Dale, A.L.; et al. Signatures of mutational processes in human cancer. Nature 2013, 500, 415–421. [Google Scholar] [CrossRef] [Green Version]
  62. Conticello, S.G. Creative deaminases, self-inflicted damage, and genome evolution. Ann. N. Y. Acad. Sci. 2012, 1267, 79–85. [Google Scholar] [CrossRef] [PubMed]
  63. Cortés-Ciriano, I.; Lee, J.J.K.; Xi, R.; Jain, D.; Jung, Y.L.; Yang, L.; Gordenin, D.; Klimczak, L.J.; Zhang, C.Z.; Pellman, D.S.; et al. Comprehensive analysis of chromothripsis in 2,658 human cancers using whole-genome sequencing. Nat. Genet. 2020, 52, 331–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Stephens, P.J.; Greenman, C.D.; Fu, B.; Yang, F.; Bignell, G.R.; Mudie, L.J.; Pleasance, E.D.; Lau, K.W.; Beare, D.; Stebbings, L.A.; et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell 2011, 144, 27–40. [Google Scholar] [CrossRef]
  65. Zhang, C.-Z.; Spektor, A.; Cornils, H.; Francis, J.M.; Jackson, E.K.; Liu, S.; Meyerson, M.; Pellman, D. Chromothripsis from DNA damage in micronuclei. Nature 2015, 522, 179–184. [Google Scholar] [CrossRef] [Green Version]
  66. Fontana, M.C.; Marconi, G.; Milosevic Feenstra, J.D.; Fonzi, E.; Papayannidis, C.; Ghelli Luserna di Rorà, A.; Padella, A.; Solli, V.; Franchini, E.; Ottaviani, M.; et al. Chromothripsis in acute myeloid leukemia: Biological features and impact on survival. Leukemia 2018. [Google Scholar] [CrossRef]
  67. Korbel, J.O.; Campbell, P.J. Criteria for inference of chromothripsis in cancer genomes. Cell 2013, 152, 1226–1236. [Google Scholar] [CrossRef] [Green Version]
  68. Maciejowski, J.; Li, Y.; Bosco, N.; Campbell, P.J.; de Lange, T. Chromothripsis and Kataegis Induced by Telomere Crisis. Cell 2015, 163, 1641–1654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Dewhurst, S.M. Chromothripsis and telomere crisis: Engines of genome instability. Curr. Opin. Genet. Dev. 2020, 60, 41–47. [Google Scholar] [CrossRef] [PubMed]
  70. Liu, P.; Erez, A.; Sreenath Nagamani, S.C.; Dhar, S.U.; Kołodziejska, K.E.; Dharmadhikari, A.V.; Cooper, M.L.; Wiszniewska, J.; Zhang, F.; Withers, M.A.; et al. Chromosome catastrophes involve replication mechanisms generating complex genomic rearrangements. Cell 2011, 146, 889–903. [Google Scholar] [CrossRef] [Green Version]
  71. Lee, J.A.; Carvalho, C.M.B.; Lupski, J.R. A DNA Replication Mechanism for Generating Nonrecurrent Rearrangements Associated with Genomic Disorders. Cell 2007, 131, 1235–1247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Hastings, P.J.; Ira, G.; Lupski, J.R. A Microhomology-Mediated Break-Induced Replication Model for the Origin of Human Copy Number Variation. PLoS Genet. 2009, 5, e1000327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Liu, S.; Kwon, M.; Mannino, M.; Yang, N.; Renda, F.; Khodjakov, A.; Pellman, D. Nuclear envelope assembly defects link mitotic errors to chromothripsis. Nature 2018, 561, 551–555. [Google Scholar] [CrossRef] [PubMed]
  74. Arlt, M.F.; Wilson, T.E.; Glover, T.W. Replication stress and mechanisms of CNV formation. Curr. Opin. Genet. Dev. 2012, 22, 204–210. [Google Scholar] [CrossRef] [Green Version]
  75. Baca, S.C.; Prandi, D.; Lawrence, M.S.; Mosquera, J.M.; Romanel, A.; Drier, Y.; Park, K.; Kitabayashi, N.; MacDonald, T.Y.; Ghandi, M.; et al. Punctuated evolution of prostate cancer genomes. Cell 2013, 153, 666–677. [Google Scholar] [CrossRef] [Green Version]
  76. Shen, M.M. Chromoplexy: A New Category of Complex Rearrangements in the Cancer Genome. Cancer Cell 2013, 23, 567–569. [Google Scholar] [CrossRef] [Green Version]
  77. Zhang, C.-Z.Z.; Leibowitz, M.L.; Pellman, D. Chromothripsis and beyond: Rapid genome evolution from complex chromosomal rearrangements. Genes Dev. 2013, 27, 2513–2530. [Google Scholar] [CrossRef] [Green Version]
  78. Pellestor, F. Chromoanagenesis: Cataclysms behind complex chromosomal rearrangements. Mol. Cytogenet. 2019, 12, 1–12. [Google Scholar] [CrossRef] [Green Version]
  79. Guo, X.; Ni, J.; Liang, Z.; Xue, J.; Fenech, M.F.; Wang, X. The molecular origins and pathophysiological consequences of micronuclei: New insights into an age-old problem. Mutat. Res.-Rev. Mutat. Res. 2019, 779, 1–35. [Google Scholar] [CrossRef]
  80. Palumbo, A.; Avet-Loiseau, H.; Oliva, S.; Lokhorst, H.M.; Goldschmidt, H.; Tosinol, L.; Richardson, P.; Caltagirone, S.; Lahuerta, J.J.; Facon, T.; et al. Revised international staging system for multiple myeloma: A report from international myeloma working group. J. Clin. Oncol. 2015, 33, 2863–2869. [Google Scholar] [CrossRef]
  81. Greipp, P.R.; Miguel, J.S.; Durie, B.G.M.; Crowley, J.J.; Barlogie, B.; Bladé, J.; Boccadoro, M.; Child, J.A.; Avet-Loiseau, H.; Kyle, R.A.; et al. International staging system for multiple myeloma. J. Clin. Oncol. 2005, 23, 3412–3420. [Google Scholar] [CrossRef] [PubMed]
  82. Jackson, G.H.; Davies, F.E.; Pawlyn, C.; Cairns, D.A.; Striha, A.; Collett, C.; Hockaday, A.; Jones, J.R.; Kishore, B.; Garg, M.; et al. Lenalidomide maintenance versus observation for patients with newly diagnosed multiple myeloma (Myeloma XI): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2019, 20, 57–73. [Google Scholar] [CrossRef] [Green Version]
  83. Jackson, G.H.; Davies, F.E.; Pawlyn, C.; Cairns, D.A.; Striha, A.; Collett, C.; Waterhouse, A.; Jones, J.R.; Kishore, B.; Garg, M.; et al. Response-adapted intensification with cyclophosphamide, bortezomib, and dexamethasone versus no intensification in patients with newly diagnosed multiple myeloma (Myeloma XI): A multicentre, open-label, randomised, phase 3 trial. Lancet Haematol. 2019, 6, e616–e629. [Google Scholar] [CrossRef] [Green Version]
  84. Perrot, A.; Lauwers-Cances, V.; Tournay, E.; Hulin, C.; Chretien, M.L.; Royer, B.; Dib, M.; Decaux, O.; Jaccard, A.; Belhadj, K.; et al. Development and validation of a cytogenetic prognostic index predicting survival in multiple myeloma. J. Clin. Oncol. 2019, 37, 1657–1665. [Google Scholar] [CrossRef]
  85. Samur, M.K.; Aktas Samur, A.; Fulciniti, M.; Szalat, R.; Han, T.; Shammas, M.; Richardson, P.; Magrangeas, F.; Minvielle, S.; Corre, J.; et al. Genome-Wide Somatic Alterations in Multiple Myeloma Reveal a Superior Outcome Group. J. Clin. Oncol. 2020, 38, JCO2000461. [Google Scholar] [CrossRef]
  86. Yip, K.; Melcher, A.; Harrington, K.; Illidge, T.; Nobes, J.; Webster, A.; Smith, D.; Lorigan, P.; Nathan, P.; Larkin, J. Pembrolizumab in Combination with Radiotherapy for Metastatic Melanoma—Introducing the PERM Trial. Clin. Oncol. 2018, 30, 201–203. [Google Scholar] [CrossRef] [Green Version]
  87. Gandhi, L.; Rodríguez-Abreu, D.; Gadgeel, S.; Esteban, E.; Felip, E.; De Angelis, F.; Domine, M.; Clingan, P.; Hochmair, M.J.; Powell, S.F.; et al. Pembrolizumab plus Chemotherapy in Metastatic Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 378, 2078–2092. [Google Scholar] [CrossRef]
  88. Paner, A.; Patel, P.; Dhakal, B. The evolving role of translocation t(11;14) in the biology, prognosis, and management of multiple myeloma. Blood Rev. 2020, 41. [Google Scholar] [CrossRef]
  89. Pinto, V.; Bergantim, R.; Caires, H.R.; Seca, H.; Guimaraes, J.E.; Vasconcelos, M.H. Multiple myeloma: Available therapies and causes of drug resistance. Cancers 2020, 12, 407. [Google Scholar] [CrossRef] [Green Version]
  90. Chng, W.J.; Dispenzieri, A.; Chim, C.S.; Fonseca, R.; Goldschmidt, H.; Lentzsch, S.; Munshi, N.; Palumbo, A.; Miguel, J.S.; Sonneveld, P.; et al. IMWG consensus on risk stratification in multiple myeloma. Leukemia 2014, 28, 269–277. [Google Scholar] [CrossRef]
  91. Boyd, K.D.; Ross, F.M.; Chiecchio, L.; Dagrada, G.P.; Konn, Z.J.; Tapper, W.J.; Walker, B.A.; Wardell, C.P.; Gregory, W.M.; Szubert, A.J.; et al. A novel prognostic model in myeloma based on co-segregating adverse FISH lesions and the ISS: Analysis of patients treated in the MRC Myeloma IX trial. Leukemia 2012, 26, 349–355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Popovic, R.; Licht, J.D. MEK and MAF in myeloma therapy. Blood 2011, 117, 2300–2302. [Google Scholar] [CrossRef] [PubMed]
  93. Avet-Loiseau, H.; Attal, M.; Moreau, P.; Charbonnel, C.; Garban, F.; Hulin, C.; Leyvraz, S.; Michallet, M.; Yakoub-Agha, I.; Garderet, L.; et al. Genetic abnormalities and survival in multiple myeloma: The experience of the Intergroupe Francophone du Myélome. Blood 2007, 109, 3489–3495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Drach, J.; Ackermann, J.; Fritz, E.; Kromer, E.; Schuster, R.; Gisslinger, H.; DeSantis, M.; Zojer, N.; Fiegl, M.; Roka, S.; et al. Presence of a p53 gene deletion in patients with multiple myeloma predicts for short survival after conventional-dose chemotherapy. Blood 1998, 92, 802–809. [Google Scholar] [CrossRef]
  95. Chang, H.; Qi, C.; Reece, D.; Stewart, A.K. P53 Gene Deletion Detected By Fluorescence in Situ Hybridization Is an Adverse Prognostic Factor for Patients With Multiple Myeloma Following Autologous Stem Cell Transplantation. Blood 2005, 105, 358–360. [Google Scholar] [CrossRef] [Green Version]
  96. Lonial, S.; Dimopoulos, M.; Palumbo, A.; White, D.; Grosicki, S.; Spicka, I.; Walter-Croneck, A.; Moreau, P.; Mateos, M.V.; Magen, H.; et al. Elotuzumab therapy for relapsed or refractory multiple myeloma. N. Engl. J. Med. 2015, 373, 621–631. [Google Scholar] [CrossRef] [Green Version]
  97. Raje, N.; Berdeja, J.; Lin, Y.; Siegel, D.; Jagannath, S.; Madduri, D.; Liedtke, M.; Rosenblatt, J.; Maus, M.V.; Turka, A.; et al. Anti-BCMA CAR T-cell therapy bb2121 in relapsed or refractory multiple myeloma. N. Engl. J. Med. 2019, 380, 1726–1737. [Google Scholar] [CrossRef]
  98. Ramos, C.A.; Savoldo, B.; Torrano, V.; Ballard, B.; Zhang, H.; Dakhova, O.; Liu, E.; Carrum, G.; Kamble, R.T.; Gee, A.P.; et al. Clinical responses with T lymphocytes targeting malignancy-associated κ light chains. J. Clin. Investig. 2016, 126, 2588–2596. [Google Scholar] [CrossRef] [Green Version]
  99. Lesokhin, A.M.; Ansell, S.M.; Armand, P.; Scott, E.C.; Halwani, A.; Gutierrez, M.; Millenson, M.M.; Cohen, A.D.; Schuster, S.J.; Lebovic, D.; et al. Nivolumab in patients with relapsed or refractory hematologic malignancy: Preliminary results of a phase ib study. J. Clin. Oncol. 2016, 34, 2698–2704. [Google Scholar] [CrossRef] [Green Version]
  100. Usmani, S.Z.; Schjesvold, F.; Oriol, A.; Karlin, L.; Cavo, M.; Rifkin, R.M.; Yimer, H.A.; LeBlanc, R.; Takezako, N.; McCroskey, R.D.; et al. Pembrolizumab plus lenalidomide and dexamethasone for patients with treatment-naive multiple myeloma (KEYNOTE-185): A randomised, open-label, phase 3 trial. Lancet Haematol. 2019, 6, e448–e458. [Google Scholar] [CrossRef]
  101. Lokhorst, H.M.; Plesner, T.; Laubach, J.P.; Nahi, H.; Gimsing, P.; Hansson, M.; Minnema, M.C.; Lassen, U.; Krejcik, J.; Palumbo, A.; et al. Targeting CD38 with daratumumab monotherapy in multiple myeloma. N. Engl. J. Med. 2015, 373, 1207–1219. [Google Scholar] [CrossRef] [PubMed]
  102. Yakoub-Agha, I.; Chabannon, C.; Bader, P.; Basak, G.W.; Bonig, H.; Ciceri, F.; Corbacioglu, S.; Duarte, R.F.; Einsele, H.; Hudecek, M.; et al. Management of adults and children undergoing chimeric antigen receptor T-cell therapy: Best practice recommendations of the European Society for Blood and Marrow Transplantation (EBMT) and the Joint Accreditation Committee of ISCT and EBMT (JACIE). Haematologica 2020, 105, 297–316. [Google Scholar] [CrossRef] [PubMed]
  103. Gay, F.; Oliva, S.; Petrucci, M.T.; Conticello, C.; Catalano, L.; Corradini, P.; Siniscalchi, A.; Magarotto, V.; Pour, L.; Carella, P.; et al. Chemotherapy plus lenalidomide versus autologous transplantation, followed by lenalidomide plus prednisone versus lenalidomide maintenance, in patients with multiple myeloma: A randomised, multicentre, phase 3 trial. Lancet Oncol. 2015, 16, 1617–1629. [Google Scholar] [CrossRef]
  104. Moreau, P.; Attal, M.; Hulin, C.; Arnulf, B.; Belhadj, K.; Benboubker, L.; Béné, M.C.; Broijl, A.; Caillon, H.; Caillot, D.; et al. Bortezomib, thalidomide, and dexamethasone with or without daratumumab before and after autologous stem-cell transplantation for newly diagnosed multiple myeloma (CASSIOPEIA): A randomised, open-label, phase 3 study. Lancet 2019, 394, 29–38. [Google Scholar] [CrossRef]
  105. Dimopoulos, M.A.; Gay, F.; Schjesvold, F.; Beksac, M.; Hajek, R.; Weisel, K.C.; Goldschmidt, H.; Maisnar, V.; Moreau, P.; Min, C.K.; et al. Oral ixazomib maintenance following autologous stem cell transplantation (TOURMALINE-MM3): A double-blind, randomised, placebo-controlled phase 3 trial. Lancet 2019, 393, 253–264. [Google Scholar] [CrossRef] [Green Version]
  106. Cavo, M.; Gay, F.; Beksac, M.; Pantani, L.; Petrucci, M.T.; Dimopoulos, M.A.; Dozza, L.; van der Holt, B.; Zweegman, S.; Oliva, S.; et al. Autologous haematopoietic stem-cell transplantation versus bortezomib–melphalan–prednisone, with or without bortezomib–lenalidomide–dexamethasone consolidation therapy, and lenalidomide maintenance for newly diagnosed multiple myeloma (EMN02/HO95): A mult. Lancet Haematol. 2020, 7, e456–e468. [Google Scholar] [CrossRef]
  107. Zweegman, S.; Van Der Holt, B.; Mellqvist, U.H.; Salomo, M.; Bos, G.M.J.; Levin, M.D.; Visser-Wisselaar, H.; Hansson, M.; van der Velden, A.W.G.; Deenik, W.; et al. Melphalan, prednisone, and lenalidomide versus melphalan, prednisone, and thalidomide in untreated multiple myeloma. Blood 2016, 127, 1109–1116. [Google Scholar] [CrossRef] [Green Version]
  108. Facon, T.; Dimopoulos, M.A.; Dispenzieri, A.; Catalano, J.V.; Belch, A.; Cavo, M.; Pinto, A.; Weisel, K.; Ludwig, H.; Bahlis, N.J.; et al. Final analysis of survival outcomes in the phase 3 FIRST trial of up-front treatment for multiple myeloma. Blood 2018, 131, 301–310. [Google Scholar] [CrossRef] [Green Version]
  109. Mateos, M.V.; Dimopoulos, M.A.; Cavo, M.; Suzuki, K.; Jakubowiak, A.; Knop, S.; Doyen, C.; Lucio, P.; Nagy, Z.; Kaplan, P.; et al. Daratumumab plus bortezomib, melphalan, and prednisone for untreated myeloma. N. Engl. J. Med. 2018, 378, 518–528. [Google Scholar] [CrossRef]
  110. Mateos, M.V.; Cavo, M.; Blade, J.; Dimopoulos, M.A.; Suzuki, K.; Jakubowiak, A.; Knop, S.; Doyen, C.; Lucio, P.; Nagy, Z.; et al. Overall survival with daratumumab, bortezomib, melphalan, and prednisone in newly diagnosed multiple myeloma (ALCYONE): A randomised, open-label, phase 3 trial. Lancet 2020, 395, 132–141. [Google Scholar] [CrossRef]
  111. Facon, T.; Lee, J.H.; Moreau, P.; Niesvizky, R.; Dimopoulos, M.; Hajek, R.; Pour, L.; Jurczyszyn, A.; Qiu, L.; Klippel, Z.; et al. Carfilzomib or bortezomib with melphalan-prednisone for transplant-ineligible patients with newly diagnosed multiple myeloma. Blood 2019, 133, 1953–1963. [Google Scholar] [CrossRef] [PubMed]
  112. Voorhees, P.M.; Kaufman, J.L.; Laubach, J.; Sborov, D.W.; Reeves, B.; Rodriguez, C.; Chari, A.; Silbermann, R.; Costa, L.J.; Anderson, L.D., Jr.; et al. Daratumumab, lenalidomide, bortezomib, and dexamethasone for transplant-eligible newly diagnosed multiple myeloma: The GRIFFIN trial. Blood 2020, 136, 936–945. [Google Scholar] [CrossRef] [PubMed]
  113. Kumar, S.K.; Jacobus, S.J.; Cohen, A.D.; Weiss, M.; Callander, N.; Singh, A.K.; Parker, T.L.; Menter, A.; Yang, X.; Parsons, B.; et al. Carfilzomib or bortezomib in combination with lenalidomide and dexamethasone for patients with newly diagnosed multiple myeloma without intention for immediate autologous stem-cell transplantation (ENDURANCE): A multicentre, open-label, phase 3, randomise. Lancet Oncol. 2020, 21, 1317–1330. [Google Scholar] [CrossRef]
Figure 1. Overview of genome aberrations in Multiple Myeloma (MM). DNA damage is a leitmotif in in multiple myeloma, both at diagnosis and relapse. Nucleotide damages (ND) and single strand breaks (SSB) cause base substitutions that alter proteins and cell function. Frequent point mutations of clinical significance occur in genes involved in DNA integrity (in blue, e.g., TP53) and in cell cycle genes (in purple, e.g., kRAS). A DNA-level complex event named kataegis—a cluster of hundreds to thousands of point mutation in less than few megabases—was occasionally described in multiple myeloma. Double strand breaks (DSB) and complex genome events (CGE) widely alter genome and chromosome structure. Other than classical karyotype events (i.e., sub-chromosome deletions and translocations), in multiple myeloma were described chromothripsis, a one-step catastrophic event in which one or few chromosome were fragmented and randomly rejoined, with casual gain and losses of DNA (blue and red in figure, respectively); chromoanasynthesis a sub-chromosomal local cluster of duplications and triplications on one single chromosome (green clusters in figure); chromoplexy, a series of chained, complex inter- and intra-chromosome translocations, involving 1 to 8 chromosomes, with frequent loss of material at breakpoints. Alterations of ploidy and translocations are the 3rd category of genome abnormalities in MM, and hyperdiploidy (HD) is the main alteration in chromosome number.
Figure 1. Overview of genome aberrations in Multiple Myeloma (MM). DNA damage is a leitmotif in in multiple myeloma, both at diagnosis and relapse. Nucleotide damages (ND) and single strand breaks (SSB) cause base substitutions that alter proteins and cell function. Frequent point mutations of clinical significance occur in genes involved in DNA integrity (in blue, e.g., TP53) and in cell cycle genes (in purple, e.g., kRAS). A DNA-level complex event named kataegis—a cluster of hundreds to thousands of point mutation in less than few megabases—was occasionally described in multiple myeloma. Double strand breaks (DSB) and complex genome events (CGE) widely alter genome and chromosome structure. Other than classical karyotype events (i.e., sub-chromosome deletions and translocations), in multiple myeloma were described chromothripsis, a one-step catastrophic event in which one or few chromosome were fragmented and randomly rejoined, with casual gain and losses of DNA (blue and red in figure, respectively); chromoanasynthesis a sub-chromosomal local cluster of duplications and triplications on one single chromosome (green clusters in figure); chromoplexy, a series of chained, complex inter- and intra-chromosome translocations, involving 1 to 8 chromosomes, with frequent loss of material at breakpoints. Alterations of ploidy and translocations are the 3rd category of genome abnormalities in MM, and hyperdiploidy (HD) is the main alteration in chromosome number.
Genes 11 01453 g001
Table 1. List of aberration deriving from genome or DNA damage in multiple myeloma, sorted by incidence or prevalence. Some digits may vary from original studies due to rounding.
Table 1. List of aberration deriving from genome or DNA damage in multiple myeloma, sorted by incidence or prevalence. Some digits may vary from original studies due to rounding.
AberrationIncidence at DiagnosisPrevalenceCitations
Single nucleotide variants
MAPK pathway (mostly kRAS, nRAS, BRAF) 40%[10,11,38]
NF-KB pathway 20%[10,11,38]
DNA-repair pathway 15%[38]
TP53 mut2–4%20–40%[27,38,39,40,41]
Chromosome-level events
1q gain 31–37% [24,42,43]
+1537% [43]
+936% [43]
+1933% [43]
+533% [43]
+333% [43]
+1125% [43]
+721% [43]
6p gain 15% [42,43]
+2112% [43]
11q gain 10% [42,43]
+X9% [43]
13q loss59–60% [24,43]
16q loss 28–35% [42,43]
1p loss 24–30% [42,43]
14q loss 23–39% [24,42,43]
8p loss 22–25% [42,43]
6q loss 21–33% [42,43]
−2218% [43]
16p loss 15% [42]
12p loss 13–15% [42,43]
−2012% [43]
del(17p)7–10%80%[40,41,43]
Hyperdyploidy
Hyperdyploidy50% [19,24,44,45,46]
Translocations
t(11;14)(q13;q32) IGH/CCND115% [19]
t(14;16)(q32;q23) IGH/MAF5% [19]
t(6;14)(p21;q32) IGH/CCND31–2% [19]
t(14;20)(q32;q12) IGH/MAFB1% [19]
translocations involving MYC15–20% [19,47,48]
Rare and complex events
Kataegis3% [19]
Chromothripsis1–24% [15,18,20]
ChromoanasynthesisAnecdotical-19%Anecdotical[15,16,22]
Chromoplexy4–11%4%[15,17,18]
Table 2. Risk stratification of MM based on karyotype abnormalities, as reported in recent clinical trials. IGH = immunoglobulin heavy chain, CCDN1 = cyclin D1, MAF = MAF BZIP transcription factor, MAFB = MAF BZIP transcription factor B.
Table 2. Risk stratification of MM based on karyotype abnormalities, as reported in recent clinical trials. IGH = immunoglobulin heavy chain, CCDN1 = cyclin D1, MAF = MAF BZIP transcription factor, MAFB = MAF BZIP transcription factor B.
Standard RiskHigh RiskUltra-High Risk
Patients not in high risk or ultra-high risk groupt(11;14)(q13;q32) IGH/CCND1More than 1 high risk lesion §
t(14;16)(q32;q23) IGH/MAF
t(14;20)(q32;q12) IGH/MAFB
del (17p)
gain (1q) §
§ exclusively in some trials [82,83].
Table 3. Outcomes in transplant eligible patients with MM that were treated at diagnosis in large phase 3 trials according their genome aberrations. On 21 November 2020, we searched on PubMed (multiple myeloma AND phase 3 NOT relapse * NOT resist *) AND Clinical Trial[ptyp] AND “last 5 years”[PDat] and we received 39 results; these results were manually curated and all the trials including transplant eligible patients from diagnosis and presenting evaluable randomized data on prognostic impact of genomic aberrations in MM are included in this table. M = melphalan, V = bortezomib, P = prednisone, Dara = daratumumab, R = lenalidomide, D = dexamethasone, C = cyclophosphamide, T = talidomide, ASCT = autologous stem cell transplant, NA = not available, sCR = stringent complete response, CR = complete remission or better, MRD- = negative measurable residual disease, NR = not reached, CI = confidence interval, f-up = follow-up, * = statistically significant between study arms in sub-cohort analysis. Descriptive sentence or number of events with median follow-up are reported whenever median PFS and/or OS are not reported for the study. Some digit may vary from the original report due to rounding.
Table 3. Outcomes in transplant eligible patients with MM that were treated at diagnosis in large phase 3 trials according their genome aberrations. On 21 November 2020, we searched on PubMed (multiple myeloma AND phase 3 NOT relapse * NOT resist *) AND Clinical Trial[ptyp] AND “last 5 years”[PDat] and we received 39 results; these results were manually curated and all the trials including transplant eligible patients from diagnosis and presenting evaluable randomized data on prognostic impact of genomic aberrations in MM are included in this table. M = melphalan, V = bortezomib, P = prednisone, Dara = daratumumab, R = lenalidomide, D = dexamethasone, C = cyclophosphamide, T = talidomide, ASCT = autologous stem cell transplant, NA = not available, sCR = stringent complete response, CR = complete remission or better, MRD- = negative measurable residual disease, NR = not reached, CI = confidence interval, f-up = follow-up, * = statistically significant between study arms in sub-cohort analysis. Descriptive sentence or number of events with median follow-up are reported whenever median PFS and/or OS are not reported for the study. Some digit may vary from the original report due to rounding.
IDAuthor (Year) Study
Median f-up
Genomic AberrationTreatmentNumber of Patients with Genomic AberrationMRD- n (%)sCR, n (%)CR, n (%)ORR, n (%)Median PFS (m)Median OS (m)
1F.M. Gay (2015) [103]high risk cytogenetic as at least one abnormality: del(17p), t(4;14), or t(14;16)CRD30 (23%)NANANANA25 m (CI NA) *NR
median f-up 52 m M200-ASCT23 (18%)NANANANA33 m (CI NA) *58 m (CI NA)
t(4;14)CRD17 (13%)NANANANANANA
M200-ASCT11 (9%)NANANANANANA
t(14;16)CRD6 (5%)NANANANANANA
M200-ASCT6 (5%)NANANANANANA
del(17p)CRD10 (8%)NANANANANANA
M200-ASCT6 (5%)NANANANANANA
2G. H. Jacskson (2019)—MYELOMA XI maintenance [82]high risk as gain (1q), t(4;14), t(14;16), t(14, 20), del(17p)R maintenance102 (22%)NANANANA54 m (CI NA) *NR
median f-up 31 m observation56 (17%)NANANANA24 m (CI NA) *NR
ultra-high risk (more than one high risk lesions)R maintenance38 (8%)NANANANA23 m (CI NA) *45 m (CI NA) *
observation23 (7%)NANANANA7 m (CI NA) *33 m (CI NA) *
t(4;14)R maintenance42 (9%)NANANANA18 events (43%) *12 events (29%)
observation24 (7%)NANANANA24 events (100%) *10 events (42%)
t(14;16)R maintenance8 (2%)NANANANA4 events (50%) 3 events (37%)
observation6 (2%)NANANANA5 events (83%) 3 events (50%)
del(17p)R maintenance25 (5%)NANANANA12 events (48%) 10 events (40%)
observation13 (4%)NANANANA10 events (77%) 7 events (54%)
gain (1q)R maintenance104 (23%)NANANANA43 events (41%) *24 events (23%)
observation58 (18%)NANANANA38 events (66%) *17 events (29%)
3G. H. Jacskson (2019)—MYELOMA XI intensification § [83]high risk as gain (1q), t(4;14), t(14;16), t(14, 20), del(17p)CVD intensification32 (25%)NANANANA31 m (CI NA) *9 events (28%)
median f-up 30 m no CVD intensification48 (37%)NANANANA17 m (CI NA) *11 events (23%)
ultra-high risk (more than one high risk lesions)CVD intensification18 (14%)NANANANA24 m (CI NA) *5 events (28%)
no CVD intensification12 (9%)NANANANA8 m (CI NA) *7 events (58%)
t(4;14)CVD intensification19 (7%)NANANANA12 events (63%) *4 events (21%) *
no CVD intensification7 (2%)NANANANA7 events (70%) *5 events (50%) *
t(14;16)CVD intensification2 (1%)NANANANA2 events (100%) 1 event (50%)
no CVD intensification2 (1%)NANANANA2 events (100%) 1 event (50%)
del(17p)CVD intensification5 (2%)NANANANA5 events (71%) *2 events (29%) *
no CVD intensification12 (4%)NANANANA12 events (86%) *8 events (57%) *
gain (1q)CVD intensification25 (9%)NANANANA25 events (63%) *12 events (30%)
no CVD intensification33 (11%)NANANANA33 events (72%) *12 events (26%)
4P. Moreau (2019)—CASSIOPEIA [104]high risk as at least one abnormality: del(17p) or t(4;14)Dara-VTD82 (15%)49 (60%)20 (24%)NANA15 events (18%) NA
median f-up 18 m VTD86 (16%)38 (44%)24 (28%)NANA22 events (26%) NA
5M.A. Dimopoulos (2019)—TOURMALINE-MM3 [105]high risk cytogenetic as at least one abnormality: del(17p), t(4;14), or t(14;16)Ixazomib maintenance61 (15%)NANANANA38 events (62%)NA
median f-up 31 m placebo54 (21%)NANANANA38 events (70%)NA
6M. Cavo (2020)—EMN02/H095 [106]high risk as at least one abnormality: t(4;14), t(14;16), del(17p) ASCT135 (25%)NANANANA37.3 m (HR 0.63, 0.46–0.88) *NR (HR 0.66, 0.45–0.99) *
Median f-up 42 m VMP intensification90 (25%)NANANANA20.3 m (HR 0.63, 0.46–0.88) *51 m (HR 0.66, 0.45–0.99) *
single ASCT42 (25%)NANANANA27 m (HR 0.59, 0.34–1.03)54.7% (HR 0.70, 0.35–1.42)
double ASCT39 (22%)NANANANA46 m (HR 0.59, 0.34–1.03)61.3% (HR 0.70, 0.35–1.42)
del(17p)ASCT64 (11%)NANANANANANR (HR 0.48, 0.27–0.86) *
VMP intensification41 (10%)NANANANANA47 m (HR 0.48, 0.27–0.86) *
single ASCT 22 (13%)NANANANA30 m (HR 0.24, 0.09–0.66) *57.1% (HR 0.30, 0.08–1.08)
double ASCT18 (10%)NANANANANR (HR 0.24, 0.09–0.66) *80.2% (HR 0.30, 0.08–1.08)
t(4;14)ASCT63 (11%)NANANANANANA
VMP intensification48 (12%)NANANANANANA
single ASCT16 (9%)NANANANANANA
double ASCT20 (11%)NANANANANANA
t(14;16)ASCT20 (4%)NANANANANANA
VMP intensification15 (4%)NANANANANANA
single ASCT7 (4%)NANANANANANA
double ASCT6 (3%)NANANANANANA
§ regardless of transplant eligibility.
Table 4. Outcomes in transplant ineligible patients with MM that were treated at diagnosis in large phase 3 trials according to their genome aberrations. On 21 November 2020, we searched on PubMed (multiple myeloma AND phase 3 NOT relapse * NOT resist *) AND Clinical Trial[ptyp] AND “last 5 years”[PDat] and we received 39 results; these results were manually curated and all the trials including transplant ineligible patients from diagnosis and presenting evaluable randomized data on prognostic impact of genomic aberrations in MM are included in this table. M = melphalan, V = bortezomib, P = prednisone, Dara = daratumumab, R = lenalidomide, D = dexamethasone, Pembro = pembrolizumab, C = cyclophosphamide, T = talidomide, K = karfilzomib, NA = not available, sCR = stringent complete response, CR = complete remission or better, MRD- = negative measurable residual disease, NR = not reached, CI = confidence interval, f-up = follow-up, * = statistically significant between study arms in sub-cohort analysis. Descriptive sentence or number of events with median follow-up are reported whenever median PFS and/or OS are not reported for the study. Some digit may vary from the original report due to rounding.
Table 4. Outcomes in transplant ineligible patients with MM that were treated at diagnosis in large phase 3 trials according to their genome aberrations. On 21 November 2020, we searched on PubMed (multiple myeloma AND phase 3 NOT relapse * NOT resist *) AND Clinical Trial[ptyp] AND “last 5 years”[PDat] and we received 39 results; these results were manually curated and all the trials including transplant ineligible patients from diagnosis and presenting evaluable randomized data on prognostic impact of genomic aberrations in MM are included in this table. M = melphalan, V = bortezomib, P = prednisone, Dara = daratumumab, R = lenalidomide, D = dexamethasone, Pembro = pembrolizumab, C = cyclophosphamide, T = talidomide, K = karfilzomib, NA = not available, sCR = stringent complete response, CR = complete remission or better, MRD- = negative measurable residual disease, NR = not reached, CI = confidence interval, f-up = follow-up, * = statistically significant between study arms in sub-cohort analysis. Descriptive sentence or number of events with median follow-up are reported whenever median PFS and/or OS are not reported for the study. Some digit may vary from the original report due to rounding.
IDAuthor (Year) Study
Median f-up
Genomic AberrationTreatmentNumber of Patients with Genomic AberrationMRD- n (%)sCR, n (%)CR, n (%)ORR, n (%)Median PFS (m)Median OS (m)
1S. Zweegman (2016)—HOVON87/NMSG18 [107]del (17p)MPT-T25 (7.9%)NANANANA20 events (80%) 11 events (44%)
median f-up 36 m MPR-R19 (6%)NANANANA18 events (95%) 10 events (52%)
t(4;14)MPT-T21 (6.6%)NANANANA20 events (95%)12 events (57%) *
MPR-R19 (6%)NANANANA17 events (89%)6 events (32%) *
1q21 gainMPT-T64 (20.1%)NANANANA49 events (77%) 29 events (45%)
MPR-R67 (21.1%)NANANANA51 events (76%) 23 events (34%)
2T. Facon (2018)—FIRST [108]high risk cytogenetic as at least one abnormality: del(17p), t(4;14), or t(14;16)RD continuous43 (17%)NANA5 (12%)33 (77%)8.429.3
median f-up 67 m RD1852 (20%)NANA7 (13%)35 (67%)17.524.3
MPT47 (19%)NANA032 (68%)14.635.5
3M.V. Mateos (2018), M.V. Mateos (2019)—ALCYONE [109,110]high risk cytogenetic as at least one abnormality: del(17p), t(4;14), or t(14;16)Dara-VMP53 (14.9%)NA10 (19%)22 (42%) *49 (92.5%) * NANR
median f-up 40 m VMP45 (12.9%)NA1 (2%)11 (24%) *33 (73.3%) *NA39.2
4T. Facon (2019)—CLARION [111]high risk cytogenetic as at least one abnormality: del(17p), t(4;14), or t(14;16)KMP54 (11.3%)NANANANAns between arms; HR 0.96 (95% CI: 0.58–1.58)NA
median f-up 27 m VMP67 (14%)NANANANA NA
5G. H. Jacskson (2019)—MYELOMA XI maintenance [82]high risk as gain (1q), t(4;14), t(14;16), t(14, 20), del(17p)R maintenance64 (27%)NANANANA16 m (CI NA) *49 m (CI NA)
median f-up 31 m observation57 (28%)NANANANA8 m (CI NA) *46 m (CI NA)
ultra-high risk (more than one high risk lesions)R maintenance15 (6%)NANANANA14 m (CI NA)46 m (CI NA)
observation7 (3%)NANANANA11 m (CI NA)39 m (CI NA)
t(4;14)R maintenance9 (4%)NANANANA7 events (78%) 3 events (34%)
observation8 (4%)NANANANA8 events (100%) 5 events (62%)
t(14;16)R maintenance13 (5%)NANANANA11 events (85%) 8 events (62%)
observation2 (1%)NANANANA1 event (50%) 1 event (50%)
del(17p)R maintenance12 (5%)NANANANA8 events (67%) 5 events (42%)
observation11 (5%)NANANANA11 events (100%) 7 events (64%)
gain (1q)R maintenance58 (24)NANANANA60 events (62%) *27 events (47%)
observation49 (24%)NANANANA46 events (94%) *23 events (47%)
6S.Z. Usmani (2019)—KEYNOTE-185 [100]high risk cytogenetic as at least one abnormality: del(17p), t(4;14), or t(14;16)Pembro-RD24 (16%)NANANANANA4 events (21%)
median f-up: 6.6 m RD10 (7%)NANANANANA0 events
7P.M. Voorhees (2020)—GRIFFIN [112]high risk cytogenetic as at least one abnormality: del(17p), t(4;14), or t(14;16)RVd14 (14.4%)53 (51%)42 (42.4%)9 (9.1%)98 (99%)NR NR
median f-up 22 m Dara-RVd16 (16.3%)21 (20.4%)31 (32%)10 (10.3%)89 (91.8%)NR NR
8S.K. Kumar (2020)—ENDURANCE [113]t(4;14)VRd44 (12%)NANANANA18.3 m (14.9-NR) NA
median f-up 9 m KRd36 (9%)NANANANA19.3 m (12.7-NR)NA
Table 5. Recommendations of cytogenetic-driven therapy in newly diagnosed multiple myeloma patients. V = bortezomib, D = dexamethasone, C = cyclophosphamide, ASCT = autologous stem cell transplant.
Table 5. Recommendations of cytogenetic-driven therapy in newly diagnosed multiple myeloma patients. V = bortezomib, D = dexamethasone, C = cyclophosphamide, ASCT = autologous stem cell transplant.
Patient PopulationRecommendationsEvidences for Recommendations
Transplant eligible patientsNo clear advantage has been demonstrated in adding daratumumab in first line therapy to bortezomib-based regimens in high risk patients.Median PFS 26% VTD vs. 18% Dara-VTD [104]
CVD intensification after induction therapy should be offered to high and ultra-high cytogenetic risk patients, including patients with gain 1q and t(14;20), not achieving at least a VGPR before proceeding to ASCT. Median PFS 31 months with CVD intensification vs. 17 months without CVD intensification [83]
Melphalan 200 mg/sqm followed by ASCT should be preferred to consolidation with CRD in high cytogenetic risk patients. Median PFS 25 months CRD vs. 33 months M200-ASCT [103]
ASCT should be offered in high cytogenetic risk patients, particularly in del(17p). Furthermore, double ASCT should be preferred to single ASCT in del(17p).Median PFS 37 m with ASCT vs. 20 m with VMP (HR 0.63, 0.46–0.88). Median OS not reached with ASCT vs. 52 m with VMP (HR 0.66, 0.45–0.99). Median PFS not reached with double ASCT vs. 30 m with single ASCT in del(17p) patients (HR 0.24, 0.09–0.66) [106]
Lenalidomide maintenance should be offered to high and ultra-high cytogenetic risk patients, including patients with gain (1q) and t(14;20).Median PFS 54 months with R maintenance vs. 24 months with observation) [82]
Transplant ineligible patientsLenalidomide based-regimen should be preferred in t(4;14) patients.Better OS; 6 deaths with MPR-R vs. 12 deaths with MPT-T [107]
Daratumumab should be added in first line therapy to bortezomib-based triplets in patients with high cytogenetic risk.ORR 92.5% in Dara-VMP vs. 73.3% in VMP [109,110]
Lenalidomide maintenance should be offered to high cytogenetic risk patients, including patients with gain (1q) and t(14;20), but benefit is not clear in ultra-high risk patients.Median PFS 16 months with R maintenance vs. 8 months with observation [82]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sessa, M.; Cavazzini, F.; Cavallari, M.; Rigolin, G.M.; Cuneo, A. A Tangle of Genomic Aberrations Drives Multiple Myeloma and Correlates with Clinical Aggressiveness of the Disease: A Comprehensive Review from a Biological Perspective to Clinical Trial Results. Genes 2020, 11, 1453. https://0-doi-org.brum.beds.ac.uk/10.3390/genes11121453

AMA Style

Sessa M, Cavazzini F, Cavallari M, Rigolin GM, Cuneo A. A Tangle of Genomic Aberrations Drives Multiple Myeloma and Correlates with Clinical Aggressiveness of the Disease: A Comprehensive Review from a Biological Perspective to Clinical Trial Results. Genes. 2020; 11(12):1453. https://0-doi-org.brum.beds.ac.uk/10.3390/genes11121453

Chicago/Turabian Style

Sessa, Mariarosaria, Francesco Cavazzini, Maurizio Cavallari, Gian Matteo Rigolin, and Antonio Cuneo. 2020. "A Tangle of Genomic Aberrations Drives Multiple Myeloma and Correlates with Clinical Aggressiveness of the Disease: A Comprehensive Review from a Biological Perspective to Clinical Trial Results" Genes 11, no. 12: 1453. https://0-doi-org.brum.beds.ac.uk/10.3390/genes11121453

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop