The ability to rearrange the germ-line DNA to generate antibody diversity is an essential prerequisite for the production of a functional repertoire. While this is essential to prevent infections, it also represents the “Achilles heal” of the B-cell lineage, occasionally leading to malignant transformation of these cells by translocation of protooncogenes into the immunoglobulin (Ig) loci. However, in evolutionary terms this is a small price to pay for a functional immune system. The study of the configuration and rearrangements of the Ig gene loci has contributed extensively to our understanding of the natural history of development of myeloma. In addition to this, the analysis of Ig gene rearrangements in B-cell neoplasms provides information about the clonal origin of the disease, prognosis, as well as providing a clinical useful tool for clonality detection and minimal residual disease monitoring. Herein, we review the data currently available on both Ig gene rearrangements and protein patterns seen in myeloma with the aim of illustrating how this knowledge has contributed to our understanding of the pathobiology of myeloma.

During early B-cell differentiation in the bone marrow (BM) the variable (V), diversity (D), and joining (J) gene segments of the immunoglobulin (Ig) genes are rearranged in an ordered fashion to generate the primary Ig repertoire. Ig heavy chain gene (IGH) rearrangement precedes Ig light chain gene rearrangement, and DH to JH joining precedes VH to DJH joining.1,2  These rearrangements are mediated by a tightly regulated enzymatic machinery involving several different proteins operating at the DNA level, which are controlled by the recombination signal sequences (RSSs) flanking each gene segment (Figure 1).3,4  The process of allelic exclusion ensures that once a functional VDJH rearrangement has been achieved, the other IGH allele is generally excluded from further recombination attempts.5  Following successful IGH recombination, the Ig light chain loci proceed to rearrange; initial attempts occur at the Ig kappa locus (IGK), and if a functional IGK rearrangement is not achieved the Ig lambda locus (IGL) undergoes recombination.6,7  Usually, rearrangements of the IGL locus are accompanied by deletion of the nonfunctional IGK rearrangements (Figure 2).7,8 

Figure 1

Schematic diagram ofIGHgene rearrangement, CSR, and SHM during B-cell development. DH to JH recombination occurs at the pro-B stage, producing an incomplete DJH rearrangement. This is followed by VH to DJH recombination at the pre-B stage that will give rise to a complete VDJH rearrangement. If this VDJH is functional, the cell will develop into a mature B-cell–expressing surface IgM (IgM+). These cells can enter the GCs where SHM and CSR occur, thus becoming VH-mutated, class-switched (eg, IgA) memory B or plasma cells. Circles preceding the CH gene segments (except δ) represent the switch regions. Dotted lines on the VH, DH, and JH segments and the variable region of the Ig protein represent hypermutated genes after the SHM process.

Figure 1

Schematic diagram ofIGHgene rearrangement, CSR, and SHM during B-cell development. DH to JH recombination occurs at the pro-B stage, producing an incomplete DJH rearrangement. This is followed by VH to DJH recombination at the pre-B stage that will give rise to a complete VDJH rearrangement. If this VDJH is functional, the cell will develop into a mature B-cell–expressing surface IgM (IgM+). These cells can enter the GCs where SHM and CSR occur, thus becoming VH-mutated, class-switched (eg, IgA) memory B or plasma cells. Circles preceding the CH gene segments (except δ) represent the switch regions. Dotted lines on the VH, DH, and JH segments and the variable region of the Ig protein represent hypermutated genes after the SHM process.

Close modal
Figure 2

Schematic diagram of the orderedIGKandIGLgene rearrangement and SHM during B-cell development. At the pre-B cell stage, after IGH rearrangements have been completed and a μ heavy chain is produced, recombination at the IGK locus occurs between a VK and a JK segment. If such rearrangement is functional, the cell can enter the GCs and undergo SHM, with the end result a mutated IgK+ memory B or plasma cell (A). However, if the IGK rearrangements at the pre-B stage produce nonfunctional Igκ chains, additional rearrangements can occur in both IGK alleles, either before entering the GC or during the GC reaction (B). We can distinguish between 2 different types of rearrangements. First, if 5′ VK and 3′ JK segments remain still available, secondary VJK rearrangements can occur, rendering a new (functional) VJK rearrangement (B1). Alternatively, deletional rearrangements via Kde can occur, with excision of the CK region as shown or with deletion of the entire JK-CK region through direct VK to Kde recombination (B2). Such deletions are normally accompanied by rearrangement of the IGL locus in 22q11 to rescue B cells from apoptosis, which can then enter the GCs undergoing affinity maturation via the SHM process (C).

Figure 2

Schematic diagram of the orderedIGKandIGLgene rearrangement and SHM during B-cell development. At the pre-B cell stage, after IGH rearrangements have been completed and a μ heavy chain is produced, recombination at the IGK locus occurs between a VK and a JK segment. If such rearrangement is functional, the cell can enter the GCs and undergo SHM, with the end result a mutated IgK+ memory B or plasma cell (A). However, if the IGK rearrangements at the pre-B stage produce nonfunctional Igκ chains, additional rearrangements can occur in both IGK alleles, either before entering the GC or during the GC reaction (B). We can distinguish between 2 different types of rearrangements. First, if 5′ VK and 3′ JK segments remain still available, secondary VJK rearrangements can occur, rendering a new (functional) VJK rearrangement (B1). Alternatively, deletional rearrangements via Kde can occur, with excision of the CK region as shown or with deletion of the entire JK-CK region through direct VK to Kde recombination (B2). Such deletions are normally accompanied by rearrangement of the IGL locus in 22q11 to rescue B cells from apoptosis, which can then enter the GCs undergoing affinity maturation via the SHM process (C).

Close modal

Assembly of a functional IgH-IgL protein complex on the cell surface, the so-called B-cell receptor (BCR), allows B cells to escape apoptosis and proceed with maturation.9  Immature yet immunocompetent B cells expressing surface Ig exit the BM environment and move to the secondary lymphoid organs. In the lymph node B cells pass through the mantle zone to reach the germinal center (GC), where affinity maturation occurs, following encounter with their cognate antigen.10  This process involves close interaction between B cells, antigen-processing dendritic cells, and T cells, allowing B cells carrying antigen-specific receptors on their surface to survive and proliferate. At this stage, 2 major molecular events occur: somatic hypermutation (SHM) and class switch recombination (CSR).11-14  SHM introduces point mutations in the rearranged Ig genes, producing high-affinity antibodies that recognize and optimally bind to foreign antigens. After initial contact with antigen, low-affinity IgM is produced, and CSR is the mechanism that changes that isotype to IgG-, IgA-, or IgE-generating specific antibodies with different functional characteristics (Figure 1).

As a result of having undergone the processes of SHM and CSR, the Ig genes in plasma cells (PCs) from myeloma patients are characterized by heavily mutated VH regions with no intraclonal variation and carry isotype-switched IGH genes (IgG or IgA).15  A significant number of myelomas (50% to 70%) also carry translocations targeting the switch regions of the IGH genes located at chromosome 14q32.16,17  These aberrant rearrangements juxtapose oncogenes into the proximity of the powerful IGH enhancers, driving abnormal expression of the translocated oncogenes. These characteristics together with the known biased usage for certain V and D gene segments establish a molecular archaeology for myeloma in which positive and negative selection processes shape the Ig repertoire prior to the acquisition of the malignant phenotype.18-20 

Heavy chain expression patterns

Recently, the International Myeloma Working Group has collected more than 10 000 cases as part of the International Staging System (ISS) study.21  In this series 60% of cases had an IgG paraprotein, 24% had IgA, 11% produced only a light chain, 3% had IgD, and 2% had biclonal or another isotype.21  It is also possible to distinguish 4 different subclasses of IgG and 2 IgA subclasses. Pooling the published data the following distribution can be seen: 68% IgG1, 17% IgG2, 11% IgG3, and 4% IgG4 for the IgG group and 93% IgA1 and 7% IgA2 for the IgA cases.22,23  These percentages largely reflect the proportions of Ig produced per day in normal individuals and, overall, they correlate with previous findings by different groups.24,25 

Myeloma has been considered to be a disease of postswitch cells, but it is now well established that preswitched IgM and IgD myelomas exist, though they are uncommon (0.5% and 2%, respectively, in the Mayo Clinic series).24  Similarly, only 39 IgE myelomas have been described to date. Despite their low frequency, IgE, IgD, IgM, and nonsecretory myelomas are of special interest due to their associations and biologic implications. A collection of 33 IgE, IgD, IgM, and nonsecretory myelomas showed an overrepresentation of t(11;14)(q13;q32) compared with IgG/IgA myelomas, with 83% of cases carrying the translocation.26 

Nonsecretory and light chain–only myelomas are seen in 1% to 3% and 10% to 20% of the total, respectively.21,24,27  These data have to be interpreted with caution because new techniques, such as the serum-free light chain assay, can actually detect portions of the Ig molecule in nonsecretory myelomas. Furthermore, to conclusively define a case of true nonsecretory myeloma, the absence of a paraprotein must be maintained, with M protein developing in up to 24% of patients during follow-up.24  Consequently, true nonsecretory myeloma may account for less than 1% of all myelomas.

Light chain expression patterns

Neoplastic PCs produce either κ or λ Ig light chain at a similar ratio to that normally produced in healthy individuals, 63% IgL-κ and 37% IgL-λ.24  Although such distribution is thought to be similar for the different myeloma subtypes, a bias toward Igλ expression has been noted in IgD-only myeloma. This could theoretically result as a consequence of receptor revision.28  Three IgDλ myelomas have been analyzed in detail, and no indication of receptor revision was found.29  The Vκ-Jκ rearrangements identified were all out-of-frame and without somatic mutations and, in support of this, a study on normal B cells reported similar observations.30  Therefore, it remains to be established why IgD myeloma has a bias for Igλ expression or why Cμ deletion, which seems to be the basis of the IgD-only phenotype, occurs preferentially in Igλ+ B cells.

Biclonal/dual IgH expression

Based on protein electrophoresis data, up to 2% of myelomas can secrete 2 different IgH isotypes or subclasses, with most being IgG+/IgA+ (53%) or IgG+/IgM+ (26%).31  The existence of true biclonal myeloma based on the presence of more that 1 IgH subtype must be carefully reviewed because both serum M components may share the same clonal origin with an identical variable region rather than being 2 independent clones. Thus, at the RNA level, clonotypic Cμ transcripts can be found in the BM of 68% of patients with IgG+ myeloma, consistent with the idea that these cells could be the clonogenic origin of the tumor clone.32,33  Similarly, in IgM+ myelomas Cγ transcripts with a clonogenic CDR3 region can be identified, consistent with the cell of origin being able to undergo the CSR process.34  Unfortunately, myelomas secreting 2 M components as detected by protein electrophoresis have not been studied in enough detail to allow us to comment on their clonal relationship. In this sense, they could also represent truly independent myeloma clones, with ongoing CSR detectable in subclones derived from the same myeloma progenitor or, alternatively, lack of allelic exclusion within the same myeloma cell.35 

Biclonal Igκ/Igλ expression

In contrast to cases showing 2 different IgH subtypes, cases characterized by 2 different IgL peaks can be considered as truly biclonal, because no molecular mechanism for changing light chain expression within the same myeloma clone has been described so far. Biclonal myeloma cases expressing both kappa and lambda are not exceptional, although its frequency has been estimated to be only around 1% to 2%.21,36  This low frequency has led to difficulty in interpreting the clinical characteristics of these patients; some authors having found no difference in the clinical characteristics and prognosis,31  while others suggest a poor response to therapy.37  Two cases expressing cytoplasmic Igκ and Igλ but secreting only IgGλ have been described by ourselves (M.v.d.B., unpublished results, June 2002), suggesting that it is possible to produce both light chain isotypes. This observation can be explained by the fact that the Igκ light chain was unable to assemble with the Ig heavy chain and illustrates how a functional IgH-IgL assembly may be one of the mechanisms leading to allelic exclusion and monospecificity.

The IGH locus contains 27 DH segments, 6 JH segments, and up to 44 functional VH gene segments.38  Rearranged VDJH genes represent the variable domain of the IgH molecules (Figure 1), with the V-D-J joining area—or complementarity-determining region (CDR)—constituting the hypervariable domain that determines the Ig specificity for a particular antigen.2 

Patterns of IGH rearrangements

Most malignant PCs secrete mature antibodies (class switched and hypermutated) coded by a functional VDJH rearrangement.1,39  Southern blot analysis of IGH rearrangements in mature B-cell malignancies shows the presence of JH rearrangements in virtually all samples, with up to 80% containing biallelic rearrangements.40  It has recently been shown by polymerase chain reaction (PCR) that up to 60% of myeloma samples contain a single DJH rearrangement on the nonproductively rearranged allele. The remaining biallelic samples usually contain 2 VDJH rearrangements, 1 of which is nonfunctional.20,41  These data are consistent with the theoretical and observed frequencies of VDJH and DJH rearrangements in normal B cells, which again supports the regulated model of allelic exclusion during B-cell development (reviewed by Jung et al42 ).

VH, DH, and JH gene segment usage

Preferential usage of Ig gene segments has been shown during the different stages of normal B-cell maturation.43,44  The VH3 family contains more members than any other and, consequently, it comprises most VH gene segments found in myeloma, followed by the VH4 and VH1 families (Table 1).18-20,45,46  VH3-30 has been consistently found to be preferentially rearranged in myeloma, followed by VH5-51, VH1-69, and VH3-23 (Table 2).18,20,46  VH3-30 and VH3-23 are the most frequently used gene segments both in normal and malignant B-cell populations,47-50  suggesting that their preferential usage is not a myeloma-specific feature but reflects the frequency seen in normal VDJH recombination. Probably the most interesting finding in this area of research is the complete absence of VH4-34 in the functional VDJH repertoire of myeloma cells,18,51  although it can be found in nonfunctional or extensively hypermutated IGH rearrangements from myeloma samples.20  The fact that VH4-34–producing cells are prevented from differentiating into antibody-producing plasma cells in healthy individuals (although not in patients with systemic lupus erythematosus)52  means that the lack of VH4-34 in the functional repertoire of myeloma cells is not a disease-specific feature but a normal B-cell differentiation mechanism of negative selection. It is well known that VH4-34 encodes antibodies that are intrinsically autoreactive, and this finding has also been proposed as an explanation for the paucity of autoimmune phenomena in myeloma. Moreover, VH4-34 is extensively represented in the normal B-cell repertoire49,52-55  and in B-cell malignancies derived from all preplasma cell stages of differentiation.56,57  Taken together, these data indicate that myeloma cells arise from a B cell in a late stage of differentiation in which negative selection of autoreactive clones has already occurred, as opposed to other B-cell malignancies, where VH4-34 is overrepresented.

Table 1

VH family usage in IGH rearrangements in multiple myeloma

VH1, no. (%)VH2, no. (%)VH3, no. (%)VH4, no. (%)VH5, no. (%)VH6, no. (%)No. of patients
Baker et al, 199445  5 (17.9) 1 (3.6) 15 (53.6) 6 (21.4) 0 (0) 1 (3.6) 28 
Rettig et al, 199618  13 (18.1) 4 (5.6) 34 (47.2) 14 (19.4) 6 (8.3) 1 (1.4) 72 
Kiyoi et al, 199619  4 (17.4) 1 (4.3) 10 (43.5) 4 (17.4) 4 (17.4) 0 (0.0) 23 
Gonzalez et al, 200520  6 (8.2) 5 (6.8) 42 (57.5) 15 (20.6) 5 (6.8) 0 (0.0) 73 
Hadzidimitriou et al, 200646  14 (18.9) 6 (8.1) 31 (41.9) 16 (21.6) 6 (8.1) 1 (1.4) 74 
Total 42 (15.6) 17 (6.3) 132 (48.9) 55 (20.4) 21 (7.8) 3 (1.1) 270 
VH1, no. (%)VH2, no. (%)VH3, no. (%)VH4, no. (%)VH5, no. (%)VH6, no. (%)No. of patients
Baker et al, 199445  5 (17.9) 1 (3.6) 15 (53.6) 6 (21.4) 0 (0) 1 (3.6) 28 
Rettig et al, 199618  13 (18.1) 4 (5.6) 34 (47.2) 14 (19.4) 6 (8.3) 1 (1.4) 72 
Kiyoi et al, 199619  4 (17.4) 1 (4.3) 10 (43.5) 4 (17.4) 4 (17.4) 0 (0.0) 23 
Gonzalez et al, 200520  6 (8.2) 5 (6.8) 42 (57.5) 15 (20.6) 5 (6.8) 0 (0.0) 73 
Hadzidimitriou et al, 200646  14 (18.9) 6 (8.1) 31 (41.9) 16 (21.6) 6 (8.1) 1 (1.4) 74 
Total 42 (15.6) 17 (6.3) 132 (48.9) 55 (20.4) 21 (7.8) 3 (1.1) 270 
Table 2

Preferential VH gene segment usage in IGH rearrangements in MM

Hadzidimitriou et al, 200646 Gonzalez et al, 200520 Rettig et al, 199618 No.* (%)
3-30 22 (10) 
5-51 12 (5.5) 
3-23 11 (5) 
1-69 11 (5) 
3-15 10 (4.6) 
3-09 9 (4.1) 
4-59 9 (4.1) 
3-11 7 (3.2) 
Total per series 33 32 26 91 (41.6) 
Hadzidimitriou et al, 200646 Gonzalez et al, 200520 Rettig et al, 199618 No.* (%)
3-30 22 (10) 
5-51 12 (5.5) 
3-23 11 (5) 
1-69 11 (5) 
3-15 10 (4.6) 
3-09 9 (4.1) 
4-59 9 (4.1) 
3-11 7 (3.2) 
Total per series 33 32 26 91 (41.6) 
*

Total = 219.

Includes the polymorphic VH segment 3-30.3.

DH3 and DH2 families are overrepresented in both the VDJH and DJH rearrangements in myeloma,19,20,46  a finding most likely reflecting normal frequencies of IGH rearrangements at the pro-B cell stage. However, some particular segments like DH1-07 are found in DJH rearrangements at normal frequencies whereas they are completely excluded from the VDJH repertoire.20,46  These data again suggest that particular gene segments can be negatively selected after VDJH recombination in precursor cells. Taking into account all of the above findings, the obvious conclusion is that environmentally encountered antigen plays at least some part in the etiology of myeloma and/or normal PC differentiation.

Somatic hypermutation

All VDJH rearrangements in myeloma are hypermutated, containing higher mutation rates than any other B-cell malignancy, with an average rate of 9%, with up to 23% mutations being seen in some samples.41,51,58,59  Several studies have indicated that the pattern of SHM in IGH rearrangements can be accounted for by antigen selection according to a binomial distribution model.60  However, about 40% of myeloma patients do not display such pattern in the IGH genes.

As opposed to VDJH rearrangements, most incomplete DJH rearrangements in myeloma are characterized by the absence of SHM.41  This finding has been applied in clinical strategies to improve detection rates aimed at demonstrating clonality and for PCR-based minimal residual disease (MRD) follow-up studies.61,62  Nonetheless, about 10% of DJH rearrangements show some evidence of mutation, containing less than 98% homology to the closest germ-line gene.41  This finding may have biologic significance or simply reflect the presence of previously unknown polymorphisms. However, the nature of this phenomenon is currently uncertain.

Molecular explanation of IgH-negative myeloma

Lack of IgH protein production in nonsecretory myeloma can theoretically be caused by abnormalities at various levels (ie, at the genomic level, at the transcriptional level, at the translational level, or through rapid degradation of newly synthesized IgH protein). In a series of 9 IgH-negative myeloma cases and cell lines investigated to address this issue, lack of IGH transcription was suggested as the major cause.63  However, in a study of 12 primary IgH-negative myeloma cases in which the IGH locus was analyzed in detail, gene rearrangement defects at the DNA level were confirmed as the principal cause. In 1 case a biallelic deletion of the JH region was found, whereas in 5 others only incomplete DJH rearrangements were observed, mostly associated with deletion of the other allele. This finding of exclusive DJH rearrangements in combination with mutated IGK genes can only be explained in terms of normal B-cell development through deletion of the functional IGH allele. Also, the use of CH probes showed that rearrangements of the JH and CH regions were not concordant in 5 cases, probably due to illegitimate CSR resulting in chromosomal translocation into the IGH locus affecting the other allele.64  Consistent with this, using fluorescence in situ hybridization (FISH) most myeloma samples and cell lines lacking IgH production have been found to contain chromosome translocations.65,66  Therefore, the available data suggest that aberrations at the DNA level are the major cause of nonsecretory myeloma.

Both the IGK and IGL loci contain a large set of V and several J gene segments but, unlike the IGH locus, there are no D gene segments (Figure 2). The human IGK light chain locus comprises many Vκ gene segments, grouped into 7 families, and 5 Jκ gene segments.67  In addition, the kappa-deleting element (Kde), located near an RSS several kilobases downstream of the Jκ-Cκ region, can rearrange to Vκ gene segments (Vκ-Kde) or to an isolated RSS in the Jκ-Cκ intron (intron-Kde).68-70  Both types of Kde gene rearrangements lead to inactivation of the IGK allele, through deletion of the entire Jκ-Cκ area or the Cκ region only, respectively (Figure 2B). The IGL locus contains many functional and pseudo Vλ gene segments, grouped into 10 families and organized in 3 large separate clusters, as well as 7 Jλ-Cλ regions, of which only the J-Cλ1, J-Cλ2, J-Cλ3, and J-Cλ7 regions are functional.71,72 

It can be anticipated that the IGK and IGL gene rearrangement patterns in Igκ+ and Igλ+ myeloma are similar to those found in other mature B-cell malignancies.46,59  Thus, Igκ+ myeloma cells harbor functional Vκ-Jκ gene rearrangements and hardly any IGL gene rearrangements, whereas Igλ+ myeloma cases show functional Vλ-Jλ gene rearrangements. Deletional rearrangements in the IGK locus are found on the nonexpressed second allele in Igκ+ myeloma cases but are even more frequent in Igλ+ cases, reflecting the end phase of nonsuccessful recombination of the IGK locus prior to IGL rearrangement (Figure 2B). In a series of 84 myeloma patients analyzed by Southern blotting, 57% of cases showed monoclonal Kde rearrangements to the Vκ/intron-RSS segments (21% monoallelic and 36% biallelic rearrangements, respectively). All patients lacking this IGK deletion and those with monoallelic deletion showed an Igκ monoclonal component in serum and/or urine, while all cases with biallelic Kde rearrangements showed an Igλ monoclonal component (36%).73 

Gene segment usage and somatic hypermutation

Several reports have produced detailed analysis of Vκ and Vλ gene segment usage in myeloma. Most of the studies show a clear preference for Vκ1 family usage (53% of the total), followed by Vκ3 (25%) (Table 3).46,59,74-78  These percentages are in line with Vκ family usage in normal B cells and other B-cell lymphoproliferative disorders, largely reflecting the distribution of Vκ gene segments in the germ-line pool. In addition, a few Vκ1 gene segments seem to be used very often; these include the Vκ1-33/1D-33, Vκ1-39/1D-39, and Vκ1-6 gene segments.59,76,78  Also, the Vκ3-20 segment, believed to be indicative of autoimmunity, was seen relatively often in 3 studies.46,59,78  This finding would seem to contrast to that seen at the IGH locus, where no evidence of VDJH rearrangements associated with autoantibodies is seen. However, the number of cases in the collective studies is too small to provide evidence for the involvement of particular (self-) antigens or epitopes in the etiology of myeloma. The studies dealing with Vλ usage in Igλ myeloma cases show largely equal Vλ1 (27%), Vλ2 (29%), and Vλ3 (41%) family usage (Table 4).46,59,77,78  Within these families, no clear preference for individual gene segments was observed.

Table 3

IGK gene segment usage in Igκ+ myeloma

StudyVκ family usage
Jκ gene segment usage
Vκ1Vκ2Vκ3Vκ4Vκ5-7Jκ1Jκ2Jκ3Jκ4Jκ5
Cannell et al, 199474  2/3 — — 1/3 — ND ND ND ND ND 
Wagner et al, 199475  3/4 — 1/4 — — 1/4 1/4 — 2/4 — 
Kosmas et al, 199676  9/11 — — 2/11 — ND ND ND ND ND 
Sahota et al, 199759  5/9 — 4/9 — — 2/9 2/9 1/9 3/9 1/9 
Kiyoi et al, 199878  8/17 3/17 4/17 1/17 1/17* 5/17 2/17 3/17 3/17 4/17 
Hadzidimitriou et al, 200646  19/42 6/42 9/42 8/42 — ND ND ND ND ND 
Total 46/86 9/59 18/72 12/73 1/17 8/30 5/30 4/30 8/30 5/30 
StudyVκ family usage
Jκ gene segment usage
Vκ1Vκ2Vκ3Vκ4Vκ5-7Jκ1Jκ2Jκ3Jκ4Jκ5
Cannell et al, 199474  2/3 — — 1/3 — ND ND ND ND ND 
Wagner et al, 199475  3/4 — 1/4 — — 1/4 1/4 — 2/4 — 
Kosmas et al, 199676  9/11 — — 2/11 — ND ND ND ND ND 
Sahota et al, 199759  5/9 — 4/9 — — 2/9 2/9 1/9 3/9 1/9 
Kiyoi et al, 199878  8/17 3/17 4/17 1/17 1/17* 5/17 2/17 3/17 3/17 4/17 
Hadzidimitriou et al, 200646  19/42 6/42 9/42 8/42 — ND ND ND ND ND 
Total 46/86 9/59 18/72 12/73 1/17 8/30 5/30 4/30 8/30 5/30 

Values are number/total number of cases.

— indicates no single case found; ND, not determined.

*

Vκ5 family usage.

Table 4

IGL gene segment usage in Igλ+ myeloma

StudyVλ family usage
Jλ gene segment usage
Vλ1Vλ2Vλ3Vλ4-10Jλ1Jλ2Jλ3Jλ4-6Jλ7
Sahota, 199759  2/6 2/6 2/6 — 1/6 3/6 — — 2/6 
Kosmas, 199877  2/6 1/6 2/6 1/6* ND ND ND ND ND 
Kiyoi, 199878  4/16 5/16 6/16 1/16 5/16 11/16 11/16 — — 
Hadzidimitriou, 200646  11/43 12/43 19/43 1/43§ 6/43 37/43 37/43 — — 
Total 19/71 20/71 29/71 3/65 6/22 14/22 14/22 0/22 2/22 
StudyVλ family usage
Jλ gene segment usage
Vλ1Vλ2Vλ3Vλ4-10Jλ1Jλ2Jλ3Jλ4-6Jλ7
Sahota, 199759  2/6 2/6 2/6 — 1/6 3/6 — — 2/6 
Kosmas, 199877  2/6 1/6 2/6 1/6* ND ND ND ND ND 
Kiyoi, 199878  4/16 5/16 6/16 1/16 5/16 11/16 11/16 — — 
Hadzidimitriou, 200646  11/43 12/43 19/43 1/43§ 6/43 37/43 37/43 — — 
Total 19/71 20/71 29/71 3/65 6/22 14/22 14/22 0/22 2/22 

Values are number/total number of cases.

— indicates no single case found; ND, not determined.

*

Vλ6 family usage.

Vλ7 family usage.

No discrimination was made between Jλ2 and Jλ3.

§

Vλ5 family usage.

Ample evidence exists for SHM in Vκ and Vλ gene segments of myeloma cases.59,74-78  However, the degree of somatic mutation can be variable between individual cases, with IgD/λ myeloma cases being renowned for extensive mutation in their VH and Vλ regions.28,29  Similar to VDJH rearrangements, there are no indications of intraclonal heterogeneity of the Vκ or Vλ gene segments, implying that there is no ongoing SHM in myeloma cells. Clustering of replacement mutations has been found in the CDR regions of both Vκ and Vλ gene segments. Probability calculations according to Chang and Casali60  using the Replacement/Silent mutation distribution revealed evidence for antigen selection in 8 of 17 (47%) myeloma cases in the studies of Kosmas et al76,77  and 4 of 15 (27%) in the study of Sahota et al.59  In the latter, it was demonstrated that if significant clustering of mutation was present, it was in either VH or VL but not both. Notably, sequence changes in the IGH and IGL CDR3, which are not considered in the binomial model, can also drastically alter antigen binding, leading to an underestimation of those Ig gene rearrangements that have been selected for by antigen.50,59,79,80  Taking together these data and the data from the IGH genes, it can be concluded that most myeloma clones have been selected for by antigen.

Jκ and Jλ gene segment usage and IGL isotype

Theoretically, the differential usage of Jκ segments has no implications for the Igκ isotype, because all Vκ-Jκ gene rearrangements will eventually use the same Cκ segment. Accordingly, published data have shown the 5 different Jκ gene segments used at similar frequencies in Igκ+ myeloma, with a slight predominance of Jκ1 and Jκ4 gene segments (Table 3).59,75,78  In the case of Igλ, however, Jλ usage has direct implications for the Cλ isotype used, as a result of the Jλ segments being located directly upstream of the respective Cλ segments. Several studies of Igλ+ myelomas by Southern blotting and PCR have shown that J-Cλ2 and, to a lesser extent, J-Cλ3 are found more frequently (Table 4).59,78,81  This Jλ2 and Jλ3 usage in myeloma cases is similar to other B-cell malignancies. Remarkably, however, myeloma cases show predominance of Jλ2 over Jλ3 usage, whereas an inverse pattern is seen in other types of B-cell malignancies.81 

Molecular explanations for Ig light chain negativity and aberrations in myeloma

Compared with IgH, the lack of Ig light chains has been studied much less extensively. Of the 3 true nonproducer cases studied in a series of 12 IgH-negative myelomas, no functional Ig light chain rearrangements could be amplified in 1 case.64  In the other 2 cases an in-frame IGK and biallelic IGL gene rearrangements could be found, suggesting that genomic aberrations may not be the major factor in Ig light chain negativity. A clue toward the unusual posttranslational modification of Ig light chains has come from a report describing a light chain disease case with a truncated Igκ chain due to unusual cleavage by signal peptidase.82  Although deletions in the V gene segments of the light chain loci have been shown to be a cause for truncated Ig light chain proteins in some cases of heavy chain disease (reviewed by Cogne et al83 ), this has not been shown to happen in myeloma, probably due to the low total number of cases so far studied.

Translocations involving the IGH locus have been reported to be the most common genetic lesion in myeloma, including t(11;14), 15% to 21%; t(4;14), 10% to 14%; t(16;14), less than 5%; and t(6;14), less than 5% of all cases.84-86  Each IGH translocation is associated with the deregulation of a specific gene (or genes) mediating ectopic expression via the influence of the powerful IGH enhancers. The t(11;14)(q13;q32) leads to the overexpression of cyclin D1,87  although it has been proposed that another gene at 11q13, MYEOV, can also be deregulated via this translocation.88  The karyotypically cryptic t(4;14)(p16;q32) involves FGFR3 and WHSC1, and it is well characterized at the molecular level.89 WHSC1 contains a SET domain and is overexpressed in t(4;14) myelomas in the form of an IGH-WHSC1 hybrid fusion transcript.89  Interestingly, the presence of IGH-WHSC1 transcripts without expression of FGFR3 is observed in a significant number (25% to 33%) of t(4:14) cases.90,91  This has been shown to occur through deletions of the whole der14 or the translocated FGFR3 gene, although different mechanisms have also been seen in some cases.90,92  The incidence of such cases (one third of all t(4;14) myelomas) and the fact that FGFR3 gene deletion is present in more than 80% of the myeloma cells in these cases suggests that it is a very early event soon after the translocation, making FGFR3 overexpression dispensable for myeloma progression/evolution. Finally, t(6;14)(p21;q32) deregulates cyclin D3 and has been reported in about 4% of the patients, whereas t(14;16)(q32;q23) deregulates c-maf and is present in about 5% of the patients. Deregulation of MYC by t(8;14)(8q24;q32) is more of a secondary event associated with tumor progression—being more commonly seen in cell lines—and is characterized by a different structure at the underlying breakpoints.93-95 

In myeloma, the breakpoints on 14q32 are localized downstream of the VDJ region, in the more centromeric switch (S) regions situated upstream of all the constant genes except δ. Physiological CSR occurs between 2 S regions producing a hybrid S region; by convention, there is breakage of DNA in Sμ and a downstream Sα or Sγ region (Figure 1). Errors in the process of breakage and reassembling the double-stranded DNA during CSR may cause DNA from another chromosome to become involved, resulting in a translocation event, so-called aberrant or illegitimate CSR. Given that most myeloma cells secrete a functional IgG or IgA molecule, any illegitimate CSR must therefore occur on the other (nonfunctional) allele. Furthermore, FISH analyses have shown that 14q32 translocations are normally present in most of the myeloma clone, indicating that they are one of the initiating events in myeloma pathogenesis.

Altogether, these data point toward defects during the physiological CSR process being responsible for the translocation events seen in most myeloma cases and that these translocations into the IGH locus provide one of the initial immortalizing events in the pathogenesis of myeloma. However, late acquisition of translocations may occur during progression of the disease (eg, t(8;14)), although in these cases the structure of the breakpoints seems to be different than in primary translocations (reviewed by Gabrea et al96 ) and begs the question about the underlying mechanisms involved in these translocations.

Any hypothesis about the etiology of myeloma should be able to explain the molecular characteristics found in myeloma cells that we have described so far: (a) evidence of antigen-driven selection with restricted VH segment usage and highly hypermutated Ig heavy and light chain genes with no intraclonal variation; (b) class-switched functional Ig molecules (mainly IgG or IgA) in most cases, with detectable preswitch (Cμ) clonotypic transcripts in some cases; and (c) aberrant Ig translocations in 40% to 70% of samples.

One such hypothesis is illustrated in Figure 3 and is further described here: during the GC reaction, mature B cells are selected by foreign antigens and undergo the SHM and CSR processes to provide a functional IgG or IgA plasma cell that subsequently migrates to the BM. At this stage, cells carrying Ig molecules specific for certain antigens (ie, VH4-34–positive cells) are excluded from becoming PCs. If illegitimate CSR occurs during the GC reaction while the cell can still undergo maturation to a memory B or plasma cell, it may exit the lymph node with an acquired ability to survive and proliferate as a consequence of oncogene deregulation. Alternatively, translocations might occur in the Ig light chain loci in 2p11 or 22q11 during the process of receptor editing.93  The acquired survival/proliferative ability would allow this premalignant clone of PCs to accumulate secondary hits, which will eventually occur in and deregulate critical genes, leading to the emergence of a malignant myeloma clone in the BM. The lack of intraclonal variation in the Ig genes of myeloma cells is consistent with this idea. A feature of this hypothesis is that the final malignant transformation events occur within the BM after the cell has abandoned the GC microenvironment. This hypothesis is similar to ideas put forward to explain the findings in follicular and mantle cell lymphoma, where Ig translocations are suggested to happen at the pro-B cell stage without altering the differentiation process to mature B cells.97  In fact, the t(14;18) is frequently detected in healthy individuals without evidence of developing follicular lymphoma.98 

Figure 3

Multiple myeloma ontogeny's hypothesis exemplified by a 14q translocation–positive case. A naive B cell harboring an incomplete DJH and a functional VDJH rearrangement enters the GC where it undergoes SHM and antigen selection (eg, selection against VH4-34 IGH rearrangements), followed by CSR. During the process of physiological CSR, double-strand breaks on the switch regions of the nonfunctional allele (DJH in the example) can be resolved by joining with a different chromosome resulting in a 14q32 translocation. If this illegitimate recombination occurs prior to legitimate CSR on the functional IGH allele, a mixture of different subpopulations (eg, IgM+ and IgG+ or IgA+) will have a survival or proliferative advantage due to the translocation, which will make them long-lived memory B or plasma cells without becoming fully malignant. After one of these subpopulations leaves the GC to become a plasma cell homing to the BM (normally IgG+ or IgA+), secondary genetic hits may occur that render such cells malignant plasma cells.

Figure 3

Multiple myeloma ontogeny's hypothesis exemplified by a 14q translocation–positive case. A naive B cell harboring an incomplete DJH and a functional VDJH rearrangement enters the GC where it undergoes SHM and antigen selection (eg, selection against VH4-34 IGH rearrangements), followed by CSR. During the process of physiological CSR, double-strand breaks on the switch regions of the nonfunctional allele (DJH in the example) can be resolved by joining with a different chromosome resulting in a 14q32 translocation. If this illegitimate recombination occurs prior to legitimate CSR on the functional IGH allele, a mixture of different subpopulations (eg, IgM+ and IgG+ or IgA+) will have a survival or proliferative advantage due to the translocation, which will make them long-lived memory B or plasma cells without becoming fully malignant. After one of these subpopulations leaves the GC to become a plasma cell homing to the BM (normally IgG+ or IgA+), secondary genetic hits may occur that render such cells malignant plasma cells.

Close modal

Switch translocations in myeloma occur preferentially in the nonfunctional allele, because only a small proportion of myeloma cases lack paraprotein expression. This suggests that at the time of the translocation, the cells are still dependent on BCR signaling, which, in turn, implies the need for a functional Ig molecule to be present at the cell surface. The presence and the molecular characteristics of the translocations in the switch regions of the IGH genes in myeloma suggest that they occur during the process of physiological CSR within the GC. On the basis of these and other observations, it can be concluded that translocation events in myeloma constitute early alterations being responsible for tumor initiation without being responsible for complete tumorigenic transformation. It is obvious, however, from gene expression and genomic studies that these translocations define subgroups of the disease with highly distinct gene signatures and outcomes.85,86,99 

The idea that myeloma-specific translocations are insufficient to produce a fully malignant phenotype is also supported by the observation that the frequency of translocations in monoclonal gammopathy of undetermined significance (MGUS) and myeloma is similar, with only a small number of MGUS patients progressing to myeloma. A possible exception is the low frequency of MGUS cases with a t(4;14), which seem to progress more rapidly to myeloma. In this context, the t(4;14) could be associated with a higher rate of genomic instability that might facilitate the acquisition of new genomic alterations leading more rapidly to myeloma.

The hypothesis put forward herein is also consistent with the experimental observations of clonotypic IgM+ VDJH rearrangements in IgG or IgA myeloma patients. The explanation is that there are residual clonotypic “preswitch” cells—which may or may not contain the Ig translocation—that have differentiated into long-lived memory B cells or PCs without becoming fully malignant (Figure 3). These cells are predicted to be dependent on BCR signaling, and the consistent and ongoing interaction of these cells with the foreign antigen they are specific for may lead to clonal expansion and recirculation of these cells through the peripheral blood and BM, allowing their detection by PCR methods. Further experimental observation that would strengthen and support these ideas would be the detection of translocations involving the switch region of the IGH genes in a small percentage of memory B and nonmalignant PCs in myeloma patients as well as in PCs from healthy individuals. However, to the best of our knowledge, such studies are yet to be published.

Studying the “molecular archaeology” of myeloma using Ig gene rearrangement and mutation status has been particularly useful because of the lack of other tools with which to examine this process. In particular, analysis of the MGUS/myeloma transition has been particularly informative. The presence of ongoing mutation within the PCs in some MGUS cases strongly suggests that the precursor cell in MGUS is continually exposed to the process of SHM and, by inference, must exist at a level still related to the GC.100  Nonetheless, it does not exclude the possibility that cells acquire mutations in their Ig regions via other routes, as yet not fully defined. What is certain, however, is that in myeloma this apparent intraclonal diversity is absent and, on relapse, most myeloma samples secrete the same paraprotein and carry the same primary chromosomal aberrations as at diagnosis, consistent with the outgrowth of a single clone of PCs located within the BM. In this sense, this PC would seem to be the logical target for treatment, and eradicating this cell should be the focus of therapeutic approaches. However, this does not fully exclude the possibility that late relapses could be caused by a myeloma “stem cell” with a different phenotype than the myeloma PC, for which new treatment strategies may prove to be more effective.

The study of Ig gene rearrangements can also help us to understand the impact of treatment on the malignant clone. The clonal Ig gene rearrangements provide specific targets to monitor the tumor cells. In this context, PCR is a sensitive tool to detect clonal rearrangements at these loci, and the lack of SHM in most DJH rearrangements increases the rate of clonality detection by PCR and offers excellent targets for monitoring MRD by consensus real-time quantitative-PCR, thus overcoming the limitations of designing specific probes for each individual patient, which is significantly more expensive and time consuming.41,61,62 

Both patients and clinicians alike are keen to understand what causes myeloma, and while classical epidemiologic approaches have established some candidate factors, there is still a lot to learn about the etiology of this disease. Studies of Ig gene rearrangements can provide insights into this process and perhaps allow us to better focus classical and molecular epidemiologic studies. In this respect, Ig gene segment usage in myeloma appears to reflect some degree of both positive and negative selection by environmentally encountered antigen. These data, together with the presence of a high degree of SHM in IGH and IGK/IGL functional rearrangements and characteristic translocations targeting the switch regions of the IGH locus in most of the myeloma cases, indicate that processes involving the Ig genes, such as BCR signaling, CSR, and SHM, play a fundamental role in early myeloma pathogenesis.

Contribution: M.v.d.B., R.G.-S., J.A.F., A.W.L., M.G., J.J.M.v.D., and J.F.S.M. wrote and reviewed the paper; and D.G. and G.J.M. designed, wrote, edited, and reviewed the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Gareth J. Morgan, Section of Haemato-Oncology, Institute of Cancer Research, 15 Cotswold Rd, Sutton, Surrey, SM2 5NG, United Kingdom; e-mail: gareth.morgan@icr.ac.uk.

1
Alt
 
FW
Yancopoulos
 
GD
Blackwell
 
TK
, et al. 
Ordered rearrangement of immunoglobulin heavy chain variable region segments.
EMBO J
1984
, vol. 
3
 (pg. 
1209
-
1219
)
2
Tonegawa
 
S
Somatic generation of antibody diversity.
Nature
1983
, vol. 
302
 (pg. 
575
-
581
)
3
Pan
 
PY
Lieber
 
MR
Teale
 
JM
The role of recombination signal sequences in the preferential joining by deletion in DH-JH recombination and in the ordered rearrangement of the IgH locus.
Int Immunol
1997
, vol. 
9
 (pg. 
515
-
522
)
4
van Zelm
 
MC
van der Burg
 
M
de Ridder
 
D
, et al. 
Ig gene rearrangement steps are initiated in early human precursor B cell subsets and correlate with specific transcription factor expression.
J Immunol
2005
, vol. 
175
 (pg. 
5912
-
5922
)
5
Chang
 
Y
Bosma
 
MJ
Bosma
 
GC
Extended duration of DH-JH rearrangement in immunoglobulin heavy chain transgenic mice: implications for regulation of allelic exclusion.
J Exp Med
1999
, vol. 
189
 (pg. 
1295
-
1305
)
6
van der Burg
 
M
Barendregt
 
BH
Szczepanski
 
T
, et al. 
Immunoglobulin light chain gene rearrangements display hierarchy in absence of selection for functionality in precursor-B-ALL.
Leukemia
2002
, vol. 
16
 (pg. 
1448
-
1453
)
7
van der Burg
 
M
Tumkaya
 
T
Boerma
 
M
, et al. 
Ordered recombination of immunoglobulin light chain genes occurs at the IGK locus but seems less strict at the IGL locus.
Blood
2001
, vol. 
97
 (pg. 
1001
-
1008
)
8
Hieter
 
PA
Korsmeyer
 
SJ
Waldmann
 
TA
Leder
 
P
Human immunoglobulin kappa light-chain genes are deleted or rearranged in lambda-producing B cells.
Nature
1981
, vol. 
290
 (pg. 
368
-
372
)
9
King
 
LB
Norvell
 
A
Monroe
 
JG
Antigen receptor-induced signal transduction imbalances associated with the negative selection of immature B cells.
J Immunol
1999
, vol. 
162
 (pg. 
2655
-
2662
)
10
MacLennan
 
IC
Germinal centers.
Annu Rev Immunol
1994
, vol. 
12
 (pg. 
117
-
139
)
11
Neuberger
 
MS
Milstein
 
C
Somatic hypermutation.
Curr Opin Immunol
1995
, vol. 
7
 (pg. 
248
-
254
)
12
Stavnezer
 
J
Antibody class switching.
Adv Immunol
1996
, vol. 
61
 (pg. 
79
-
146
)
13
Klein
 
U
Goossens
 
T
Fischer
 
M
, et al. 
Somatic hypermutation in normal and transformed human B cells.
Immunol Rev
1998
, vol. 
162
 (pg. 
261
-
280
)
14
Honjo
 
T
Kinoshita
 
K
Muramatsu
 
M
Molecular mechanism of class switch recombination: linkage with somatic hypermutation.
Annu Rev Immunol
2002
, vol. 
20
 (pg. 
165
-
196
)
15
Bakkus
 
MH
Heirman
 
C
van Riet
 
I
van Camp
 
B
Thielemans
 
K
Evidence that multiple myeloma Ig heavy chain VDJ genes contain somatic mutations but show no intraclonal variation.
Blood
1992
, vol. 
80
 (pg. 
2326
-
2335
)
16
Bergsagel
 
PL
Chesi
 
M
Nardini
 
E
, et al. 
Promiscuous translocations into immunoglobulin heavy chain switch regions in multiple myeloma.
Proc Natl Acad Sci U S A
1996
, vol. 
93
 (pg. 
13931
-
13936
)
17
Pratt
 
G
Fenton
 
JA
Proffitt
 
JA
, et al. 
True spectrum of 14q32 translocations in multiple myeloma.
Br J Haematol
1998
, vol. 
103
 (pg. 
1209
-
1210
)
18
Rettig
 
MB
Vescio
 
RA
Cao
 
J
, et al. 
VH gene usage is multiple myeloma: complete absence of the VH4.21 (VH4–34) gene.
Blood
1996
, vol. 
87
 (pg. 
2846
-
2852
)
19
Kiyoi
 
H
Naito
 
K
Ohno
 
R
Naoe
 
T
Comparable gene structure of the immunoglobulin heavy chain variable region between multiple myeloma and normal bone marrow lymphocytes.
Leukemia
1996
, vol. 
10
 (pg. 
1804
-
1812
)
20
Gonzalez
 
D
Gonzalez
 
M
Balanzategui
 
A
, et al. 
Molecular characteristics and gene segment usage in IGH gene rearrangements in multiple myeloma.
Haematologica
2005
, vol. 
90
 (pg. 
906
-
913
)
21
Greipp
 
PR
San Miguel
 
JF
Durie
 
BG
, et al. 
International staging system for multiple myeloma.
J Clin Oncol
2005
, vol. 
23
 (pg. 
3412
-
3420
)
22
Papadea
 
C
Reimer
 
CB
Check
 
IJ
IgG subclass distribution in patients with multiple myeloma or with monoclonal gammopathy of undetermined significance.
Ann Clin Lab Sci
1989
, vol. 
19
 (pg. 
27
-
37
)
23
Fasullo
 
FJ
Fritsche
 
HA
Liu
 
FJ
Hamilton
 
RG
IgG heavy-chain subclass typing of myeloma paraproteins by isoelectric focusing immunoblot analysis.
Clin Chem
1989
, vol. 
35
 (pg. 
364
-
368
)
24
Kyle
 
RA
Gertz
 
MA
Witzig
 
TE
, et al. 
Review of 1027 patients with newly diagnosed multiple myeloma.
Mayo Clin Proc
2003
, vol. 
78
 (pg. 
21
-
33
)
25
Garcia-Sanz
 
R
Gonzalez-Fraile
 
MI
Mateo
 
G
, et al. 
Proliferative activity of plasma cells is the most relevant prognostic factor in elderly multiple myeloma patients.
Int J Cancer
2004
, vol. 
112
 (pg. 
884
-
889
)
26
Avet-Loiseau
 
H
Garand
 
R
Lode
 
L
Harousseau
 
JL
Bataille
 
R
Translocation t(11;14)(q13;q32) is the hallmark of IgM, IgE, and nonsecretory multiple myeloma variants.
Blood
2003
, vol. 
101
 (pg. 
1570
-
1571
)
27
Garcia-Sanz
 
R
Orfao
 
A
Gonzalez
 
M
, et al. 
Primary plasma cell leukemia: clinical, immunophenotypic, DNA ploidy, and cytogenetic characteristics.
Blood
1999
, vol. 
93
 (pg. 
1032
-
1037
)
28
Arpin
 
C
de Bouteiller
 
O
Razanajaona
 
D
, et al. 
The normal counterpart of IgD myeloma cells in germinal center displays extensively mutated IgVH gene, Cmu-Cdelta switch, and lambda light chain expression.
J Exp Med
1998
, vol. 
187
 (pg. 
1169
-
1178
)
29
van der
 
BM
Bende
 
RJ
Aarts
 
WM
, et al. 
Biased Iglambda expression in hypermutated IgD multiple myelomas does not result from receptor revision.
Leukemia
2002
, vol. 
16
 (pg. 
1358
-
1361
)
30
Goossens
 
T
Brauninger
 
A
Klein
 
U
Kuppers
 
R
Rajewsky
 
K
Receptor revision plays no major role in shaping the receptor repertoire of human memory B cells after the onset of somatic hypermutation.
Eur J Immunol
2001
, vol. 
31
 (pg. 
3638
-
3648
)
31
Kyle
 
RA
Robinson
 
RA
Katzmann
 
JA
The clinical aspects of biclonal gammopathies. Review of 57 cases.
Am J Med
1981
, vol. 
71
 (pg. 
999
-
1008
)
32
Bakkus
 
MH
van Riet
 
I
van Camp
 
B
Thielemans
 
K
Evidence that the clonogenic cell in multiple myeloma originates from a pre-switched but somatically mutated B cell.
Br J Haematol
1994
, vol. 
87
 (pg. 
68
-
74
)
33
Reiman
 
T
Seeberger
 
K
Taylor
 
BJ
, et al. 
Persistent preswitch clonotypic myeloma cells correlate with decreased survival: evidence for isotype switching within the myeloma clone.
Blood
2001
, vol. 
98
 (pg. 
2791
-
2799
)
34
Sahota
 
SS
Garand
 
R
Mahroof
 
R
, et al. 
V(H) gene analysis of IgM-secreting myeloma indicates an origin from a memory cell undergoing isotype switch events.
Blood
1999
, vol. 
94
 (pg. 
1070
-
1076
)
35
Rassenti
 
LZ
Kipps
 
TJ
Lack of allelic exclusion in B cell chronic lymphocytic leukemia.
J Exp Med
1997
, vol. 
185
 (pg. 
1435
-
1445
)
36
Osterborg
 
A
Mellstedt
 
H
Monoclonal and biclonal immunoglobulin-producing disorders.
Eur J Haematol Suppl
1989
, vol. 
51
 (pg. 
11
-
18
)
37
Smith
 
L
Barlogie
 
B
Alexanian
 
R
Biclonal and hypodiploid multiple myeloma.
Am J Med
1986
, vol. 
80
 (pg. 
841
-
843
)
38
Matsuda
 
F
Ishii
 
K
Bourvagnet
 
P
, et al. 
The complete nucleotide sequence of the human immunoglobulin heavy chain variable region locus.
J Exp Med
1998
, vol. 
188
 (pg. 
2151
-
2162
)
39
Schlissel
 
M
Allelic exclusion of immunoglobulin gene rearrangement and expression: why and how?
Semin Immunol
2002
, vol. 
14
 (pg. 
207
-
212
)
40
Beishuizen
 
A
Verhoeven
 
MA
Mol
 
EJ
, et al. 
Detection of immunoglobulin heavy-chain gene rearrangements by Southern blot analysis: recommendations for optimal results.
Leukemia
1993
, vol. 
7
 (pg. 
2045
-
2053
)
41
Gonzalez
 
D
Balanzategui
 
A
Garcia-Sanz
 
R
, et al. 
Incomplete DJH rearrangements of the IgH gene are frequent in multiple myeloma patients: immunobiological characteristics and clinical implications.
Leukemia
2003
, vol. 
17
 (pg. 
1398
-
1403
)
42
Jung
 
D
Giallourakis
 
C
Mostoslavsky
 
R
Alt
 
FW
Mechanism and control of V(D)J recombination at the immunoglobulin heavy chain locus.
Annu Rev Immunol
2006
, vol. 
24
 (pg. 
541
-
570
)
43
Yamada
 
M
Wasserman
 
R
Reichard
 
BA
, et al. 
Preferential utilization of specific immunoglobulin heavy chain diversity and joining segments in adult human peripheral blood B lymphocytes.
J Exp Med
1991
, vol. 
173
 (pg. 
395
-
407
)
44
Wasserman
 
R
Ito
 
Y
Galili
 
N
, et al. 
The pattern of joining (JH) gene usage in the human IgH chain is established predominantly at the B precursor cell stage.
J Immunol
1992
, vol. 
149
 (pg. 
511
-
516
)
45
Baker
 
BW
Deane
 
M
Gilleece
 
MH
, et al. 
Distinctive features of immunoglobulin heavy chain variable region gene rearrangement in multiple myeloma.
Leuk Lymphoma
1994
, vol. 
14
 (pg. 
291
-
301
)
46
Hadzidimitriou
 
A
Stamatopoulos
 
K
Belessi
 
C
, et al. 
Immunoglobulin genes in multiple myeloma: expressed and non-expressed repertoires, heavy and light chain pairings and somatic mutation patterns in a series of 101 cases.
Haematologica
2006
, vol. 
91
 (pg. 
781
-
787
)
47
Brezinschek
 
HP
Brezinschek
 
RI
Lipsky
 
PE
Analysis of the heavy chain repertoire of human peripheral B cells using single-cell polymerase chain reaction.
J Immunol
1995
, vol. 
155
 (pg. 
190
-
202
)
48
Cook
 
GP
Tomlinson
 
IM
The human immunoglobulin VH repertoire.
Immunol Today
1995
, vol. 
16
 (pg. 
237
-
242
)
49
Brezinschek
 
HP
Foster
 
SJ
Brezinschek
 
RI
, et al. 
Analysis of the human VH gene repertoire. Differential effects of selection and somatic hypermutation on human peripheral CD5(+)/IgM+ and CD5(-)/IgM+ B cells.
J Clin Invest
1997
, vol. 
99
 (pg. 
2488
-
2501
)
50
Fais
 
F
Ghiotto
 
F
Hashimoto
 
S
, et al. 
Chronic lymphocytic leukemia B cells express restricted sets of mutated and unmutated antigen receptors.
J Clin Invest
1998
, vol. 
102
 (pg. 
1515
-
1525
)
51
Vescio
 
RA
Cao
 
J
Hong
 
CH
, et al. 
Myeloma Ig heavy chain V region sequences reveal prior antigenic selection and marked somatic mutation but no intraclonal diversity.
J Immunol
1995
, vol. 
155
 (pg. 
2487
-
2497
)
52
Pugh-Bernard
 
AE
Silverman
 
GJ
Cappione
 
AJ
, et al. 
Regulation of inherently autoreactive VH4–34 B cells in the maintenance of human B cell tolerance.
J Clin Invest
2001
, vol. 
108
 (pg. 
1061
-
1070
)
53
Chapman
 
CJ
Mockridge
 
CI
Hamblin
 
TJ
Stevenson
 
FK
Tracking of the V4–34 (VH4–21) gene in human tonsil reveals clonal isotype switch events and a highly variable degree of somatic hypermutation.
Clin Exp Immunol
1996
, vol. 
105
 (pg. 
360
-
368
)
54
Friedman
 
DF
Kraj
 
P
Silberstein
 
LE
VH4.21 expression in the normal human B-cell repertoire.
Ann N Y Acad Sci
1995
, vol. 
764
 (pg. 
285
-
292
)
55
Kraj
 
P
Friedman
 
DF
Stevenson
 
F
Silberstein
 
LE
Evidence for the overexpression of the VH4–34 (VH4.21) Ig gene segment in the normal adult human peripheral blood B cell repertoire.
J Immunol
1995
, vol. 
154
 (pg. 
6406
-
6420
)
56
Deane
 
M
Baker
 
BW
Norton
 
JD
Immunoglobulin VH4 gene usage in B lymphoid leukaemias.
Br J Haematol
1993
, vol. 
84
 (pg. 
242
-
249
)
57
Hsu
 
FJ
Levy
 
R
Preferential use of the VH4 Ig gene family by diffuse large-cell lymphoma.
Blood
1995
, vol. 
86
 (pg. 
3072
-
3082
)
58
Kosmas
 
C
Stamatopoulos
 
K
Stavroyianni
 
N
, et al. 
Origin and diversification of the clonogenic cell in multiple myeloma: lessons from the immunoglobulin repertoire.
Leukemia
2000
, vol. 
14
 (pg. 
1718
-
1726
)
59
Sahota
 
SS
Leo
 
R
Hamblin
 
TJ
Stevenson
 
FK
Myeloma VL and VH gene sequences reveal a complementary imprint of antigen selection in tumor cells.
Blood
1997
, vol. 
89
 (pg. 
219
-
226
)
60
Chang
 
B
Casali
 
P
The CDR1 sequences of a major proportion of human germline Ig VH genes are inherently susceptible to amino acid replacement.
Immunol Today
1994
, vol. 
15
 (pg. 
367
-
373
)
61
Gonzalez
 
D
Gonzalez
 
M
Alonso
 
ME
, et al. 
Incomplete DJH rearrangements as a novel tumor target for minimal residual disease quantitation in multiple myeloma using real-time PCR.
Leukemia
2003
, vol. 
17
 (pg. 
1051
-
1057
)
62
Sarasquete
 
ME
Garcia-Sanz
 
R
Gonzalez
 
D
, et al. 
Minimal residual disease monitoring in multiple myeloma: a comparison between allelic-specific oligonucleotide real-time quantitative polymerase chain reaction and flow cytometry.
Haematologica
2005
, vol. 
90
 (pg. 
1365
-
1372
)
63
Sakai
 
A
Kawano
 
MM
Tanabe
 
O
Kuramoto
 
A
A possible mechanism of inability of immunoglobulin heavy-chain production in Bence-Jones type myeloma cells.
Int J Hematol
1993
, vol. 
59
 (pg. 
31
-
40
)
64
Szczepanski
 
T
van tVeer
 
MB
Wolvers-Tettero
 
IL
Langerak
 
AW
van Dongen
 
JM
Molecular features responsible for the absence of immunoglobulin heavy chain protein synthesis in an IgH(-) subgroup of multiple myeloma.
Blood
2000
, vol. 
96
 (pg. 
1087
-
1093
)
65
Kuipers
 
J
Vaandrager
 
JW
Weghuis
 
DO
, et al. 
Fluorescence in situ hybridization analysis shows the frequent occurrence of 14q32.3 rearrangements with involvement of immunoglobulin switch regions in myeloma cell lines.
Cancer Genet Cytogenet
1999
, vol. 
109
 (pg. 
99
-
107
)
66
Magrangeas
 
F
Cormier
 
ML
Descamps
 
G
, et al. 
Light-chain only multiple myeloma is due to the absence of functional (productive) rearrangement of the IgH gene at the DNA level.
Blood
2004
, vol. 
103
 (pg. 
3869
-
3875
)
67
Weichhold
 
GM
Ohnheiser
 
R
Zachau
 
HG
The human immunoglobulin kappa locus consists of two copies that are organized in opposite polarity.
Genomics
1993
, vol. 
16
 (pg. 
503
-
511
)
68
Siminovitch
 
KA
Bakhshi
 
A
Goldman
 
P
Korsmeyer
 
SJ
A uniform deleting element mediates the loss of kappa genes in human B cells.
Nature
1985
, vol. 
316
 (pg. 
260
-
262
)
69
Beishuizen
 
A
Verhoeven
 
MA
Mol
 
EJ
van Dongen
 
JJ
Detection of immunoglobulin kappa light-chain gene rearrangement patterns by Southern blot analysis.
Leukemia
1994
, vol. 
8
 (pg. 
2228
-
2236
)
70
Langerak
 
AW
van Dongen
 
JJ
Recombination in the human IGK locus.
Crit Rev Immunol
2006
, vol. 
26
 (pg. 
23
-
42
)
71
Vasicek
 
TJ
Leder
 
P
Structure and expression of the human immunoglobulin lambda genes.
J Exp Med
1990
, vol. 
172
 (pg. 
609
-
620
)
72
Frippiat
 
JP
Williams
 
SC
Tomlinson
 
IM
, et al. 
Organization of the human immunoglobulin lambda light-chain locus on chromosome 22q11.2.
Hum Mol Genet
1995
, vol. 
4
 (pg. 
983
-
991
)
73
Garcia-Sanz
 
R
Lopez-Perez
 
R
Langerak
 
AW
, et al. 
Heteroduplex PCR analysis of rearranged immunoglobulin genes for clonality assessment in multiple myeloma.
Haematologica
1999
, vol. 
84
 (pg. 
328
-
335
)
74
Cannell
 
PK
Amlot
 
P
Attard
 
M
Hoffbrand
 
AV
Foroni
 
L
Variable kappa gene rearrangement in lymphoproliferative disorders: an analysis of V kappa gene usage, VJ joining and somatic mutation.
Leukemia
1994
, vol. 
8
 (pg. 
1139
-
1145
)
75
Wagner
 
SD
Martinelli
 
V
Luzzatto
 
L
Similar patterns of V kappa gene usage but different degrees of somatic mutation in hairy cell leukemia, prolymphocytic leukemia, Waldenstrom's macroglobulinemia, and myeloma.
Blood
1994
, vol. 
83
 (pg. 
3647
-
3653
)
76
Kosmas
 
C
Viniou
 
NA
Stamatopoulos
 
K
, et al. 
Analysis of the kappa light chain variable region in multiple myeloma.
Br J Haematol
1996
, vol. 
94
 (pg. 
306
-
317
)
77
Kosmas
 
C
Stamatopoulos
 
K
Papadaki
 
T
, et al. 
Somatic hypermutation of immunoglobulin variable region genes: focus on follicular lymphoma and multiple myeloma.
Immunol Rev
1998
, vol. 
162
 (pg. 
281
-
292
)
78
Kiyoi
 
H
Naito
 
K
Ohno
 
R
Saito
 
H
Naoe
 
T
Characterization of the immunoglobulin light chain variable region gene expressed in multiple myeloma.
Leukemia
1998
, vol. 
12
 (pg. 
601
-
609
)
79
Allen
 
D
Simon
 
T
Sablitzky
 
F
Rajewsky
 
K
Cumano
 
A
Antibody engineering for the analysis of affinity maturation of an anti-hapten response.
EMBO J
1988
, vol. 
7
 (pg. 
1995
-
2001
)
80
Katz
 
JB
Limpanasithikul
 
W
Diamond
 
B
Mutational analysis of an autoantibody: differential binding and pathogenicity.
J Exp Med
1994
, vol. 
180
 (pg. 
925
-
932
)
81
Tumkaya
 
T
van der Burg
 
M
Garcia-Sanz
 
R
, et al. 
Immunoglobulin lambda isotype gene rearrangements in B cell malignancies.
Leukemia
2001
, vol. 
15
 (pg. 
121
-
127
)
82
Decourt
 
C
Cogne
 
M
Rocca
 
A
Structural peculiarities of a truncated V kappa III immunoglobulin light chain in myeloma with light chain deposition disease.
Clin Exp Immunol
1996
, vol. 
106
 (pg. 
357
-
361
)
83
Cogne
 
M
Preud'homme
 
JL
Guglielmi
 
P
Immunoglobulin gene alterations in human heavy chain diseases.
Res Immunol
1989
, vol. 
140
 (pg. 
487
-
502
)
84
Chesi
 
M
Kuehl
 
WM
Bergsagel
 
PL
Recurrent immunoglobulin gene translocations identify distinct molecular subtypes of myeloma.
Ann Oncol
2000
, vol. 
11
 
suppl 1
(pg. 
131
-
135
)
85
Bergsagel
 
PL
Kuehl
 
WM
Molecular pathogenesis and a consequent classification of multiple myeloma.
J Clin Oncol
2005
, vol. 
23
 (pg. 
6333
-
6338
)
86
Avet-Loiseau
 
H
Attal
 
M
Moreau
 
P
, et al. 
Genetic abnormalities and survival in multiple myeloma: the experience of the Intergroupe Francophone du Myelome.
Blood
2007
, vol. 
109
 (pg. 
3489
-
3495
)
87
Chesi
 
M
Bergsagel
 
PL
Brents
 
LA
, et al. 
Dysregulation of cyclin D1 by translocation into an IgH gamma switch region in two multiple myeloma cell lines.
Blood
1996
, vol. 
88
 (pg. 
674
-
681
)
88
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
, vol. 
95
 (pg. 
2691
-
2698
)
89
Chesi
 
M
Nardini
 
E
Lim
 
RS
, et al. 
The t(4;14) translocation in myeloma dysregulates both FGFR3 and a novel gene, MMSET, resulting in IgH/MMSET hybrid transcripts.
Blood
1998
, vol. 
92
 (pg. 
3025
-
3034
)
90
Santra
 
M
Zhan
 
F
Tian
 
E
Barlogie
 
B
Shaughnessy
 
J
A subset of multiple myeloma harboring the t(4;14)(p16;q32) translocation lacks FGFR3 expression but maintains an IGH/MMSET fusion transcript.
Blood
2003
, vol. 
101
 (pg. 
2374
-
2376
)
91
Keats
 
JJ
Reiman
 
T
Maxwell
 
CA
, et al. 
In multiple myeloma, t(4;14)(p16;q32) is an adverse prognostic factor irrespective of FGFR3 expression.
Blood
2003
, vol. 
101
 (pg. 
1520
-
1529
)
92
Keats
 
JJ
Reiman
 
T
Belch
 
AR
Pilarski
 
LM
Ten years and counting: so what do we know about t(4;14)(p16;q32) multiple myeloma.
Leuk Lymphoma
2006
, vol. 
47
 (pg. 
2289
-
2300
)
93
Chesi
 
M
Bergsagel
 
PL
Shonukan
 
OO
, et al. 
Frequent dysregulation of the c-maf proto-oncogene at 16q23 by translocation to an Ig locus in multiple myeloma.
Blood
1998
, vol. 
91
 (pg. 
4457
-
4463
)
94
Shou
 
Y
Martelli
 
ML
Gabrea
 
A
, et al. 
Diverse karyotypic abnormalities of the c-myc locus associated with c-myc dysregulation and tumor progression in multiple myeloma.
Proc Natl Acad Sci U S A
2000
, vol. 
97
 (pg. 
228
-
233
)
95
Shaughnessy
 
J
Gabrea
 
A
Qi
 
Y
, et al. 
Cyclin D3 at 6p21 is dysregulated by recurrent chromosomal translocations to immunoglobulin loci in multiple myeloma.
Blood
2001
, vol. 
98
 (pg. 
217
-
223
)
96
Gabrea
 
A
Leif
 
BP
Michael
 
KW
Distinguishing primary and secondary translocations in multiple myeloma.
DNA Repair (Amst)
2006
, vol. 
5
 (pg. 
1225
-
1233
)
97
Tsujimoto
 
Y
Gorham
 
J
Cossman
 
J
Jaffe
 
E
Croce
 
CM
The t(14;18) chromosome translocations involved in B-cell neoplasms result from mistakes in VDJ joining.
Science
1985
, vol. 
229
 (pg. 
1390
-
1393
)
98
Schmitt
 
C
Balogh
 
B
Grundt
 
A
, et al. 
The bcl-2/IgH rearrangement in a population of 204 healthy individuals: occurrence, age and gender distribution, breakpoints, and detection method validity.
Leuk Res
2006
, vol. 
30
 (pg. 
745
-
750
)
99
Davies
 
FE
Dring
 
AM
Li
 
C
, et al. 
Insights into the multistep transformation of MGUS to myeloma using microarray expression analysis.
Blood
2003
, vol. 
102
 (pg. 
4504
-
4511
)
100
Zojer
 
N
Ludwig
 
H
Fiegl
 
M
Stevenson
 
FK
Sahota
 
SS
Patterns of somatic mutations in VH genes reveal pathways of clonal transformation from MGUS to multiple myeloma.
Blood
2003
, vol. 
101
 (pg. 
4137
-
4139
)
Sign in via your Institution