Next Article in Journal
Whole-Gene Deletions of FZD4 Cause Familial Exudative Vitreoretinopathy
Next Article in Special Issue
T-Cell Acute Lymphoblastic Leukemia: Biomarkers and Their Clinical Usefulness
Previous Article in Journal
Building a Flower: The Influence of Cell Wall Composition on Flower Development and Reproduction
Previous Article in Special Issue
Genetic Alterations and Therapeutic Targeting of Philadelphia-Like Acute Lymphoblastic Leukemia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immune Gene Rearrangements: Unique Signatures for Tracing Physiological Lymphocytes and Leukemic Cells

1
Unit for Hematological Diagnostics, Department of Internal Medicine II, University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
2
Department of Internal Medicine II, University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
*
Author to whom correspondence should be addressed.
Genes 2021, 12(7), 979; https://doi.org/10.3390/genes12070979
Submission received: 31 May 2021 / Accepted: 25 June 2021 / Published: 27 June 2021
(This article belongs to the Special Issue Genetics and Genomics of Blood Disorders)

Abstract

:
The tremendous diversity of the human immune repertoire, fundamental for the defense against highly heterogeneous pathogens, is based on the ingenious mechanism of immune gene rearrangements. Rearranged immune genes encoding the immunoglobulins and T-cell receptors and thus determining each lymphocyte’s antigen specificity are very valuable molecular markers for tracing malignant or physiological lymphocytes. One of their most significant applications is tracking residual leukemic cells in patients with lymphoid malignancies. This so called ‘minimal residual disease’ (MRD) has been shown to be the most important prognostic factor across various leukemia subtypes and has therefore been given enormous attention. Despite the current rapid development of the molecular methods, the classical real-time PCR based approach is still being regarded as the standard method for molecular MRD detection due to the cumbersome standardization of the novel approaches currently in progress within the EuroMRD and EuroClonality NGS Consortia. Each of the molecular methods, however, poses certain benefits and it is therefore expectable that none of the methods for MRD detection will clearly prevail over the others in the near future.

1. Immunoglobulin and T-Cell Receptor Rearrangements

The remarkable ability of the human immune system to recognize and eradicate the enormous number of various antigens is based on the immune receptors [1]; the surface of T-lymphocytes is covered with the T-cell receptors (TR; TRαβ or TRγδ) and B-lymphocytes produce secreted or surface-bound immunoglobulins (IG). The tremendous diversity of the immune receptor variable domains is crucial for the specific molecular recognition of virtually any antigen [2]. Such a high degree of diversity is generated by combination of a limited number of gene segments. During this so-called somatic recombination, a DNA sequence that is unique for each lymphocyte is produced [3]. Hence, each lymphocyte bears many copies of the antigen receptor with a unique variable region that determines its antigen specificity.

1.1. Structure of the Immune Receptors

Both surface-bound and secreted immunoglobulins consist of two heavy chains (IGH) and two light chains (IGκ or IGλ), which are connected by a disulphide bond. The IGH gene complex consists of V (“variable”) segments at the 5’-end, which are followed by a group of D (“diversity”) segments and 6 short J (“joining”) segments. The gene segments for the constant (C) part of the heavy chain are localized at the 3′-end of the gene complex and are responsible for the immunoglobulin class determination. IG light chains are either encoded by a kappa (IGK) or by a lambda (IGL) rearrangement. The structure of these complexes resembles the structure of the IGH rearrangements but is composed of less V and J segments and does not contain any D segments.
TR molecules are also composed of two chains, connected by a disulphide bond. The “classical” type of TR contains (TRα) and (TRβ) chains, whereas the “alternative” type contains (TRγ) and (TRδ) chains [4,5]. The variable domains of TRβ and TRδ contain all three types of gene segments (V, D, and J). TRα and TRγ chains lack D segments, similarly to immunoglobulin light chains. The gene complex for TRδ is localized within the gene complex for TRα (between the V and J segments). Any V-J rearrangement of the TRA gene segments therefore results in the loss of TRD gene segments, which means that complete TRA and TRD gene rearrangements can never be present on a single allele simultaneously (with the exception of combined TRDV-TRAJ rearrangements [6]).

1.2. Somatic (V-D-J) Recombination

Somatic recombination occurs in immature lymphocytes in primary lymphoid organs (bone marrow for B-lymphocytes, thymus for T lymphocytes). V, D, and J gene segments of IG and TR genes are rearranged and a DNA sequence, which is unique for each lymphocyte, is produced. V-D-J recombination of IGH and TRB genes is a two-step process starting with a D-J recombination followed by a V-D recombination. The recombination of the TRD locus starts with D-D recombination and continues with V-D and D-J recombinations. Genes for Igκ, Igλ, TRα, and TRγ are produced by a one-step V-J recombination. During each recombination a random number of gene segments is excised and the remaining segments are joined together. This gives rise to tens of millions of possible combinations of V, (D) and J segments.
This process occurs in parallel on both chromosomes. As soon as a productive rearrangement is formed, it is transcribed into mRNA. At that point, the recombination process on the second chromosome is stopped and therefore only one type of antigen receptor is produced by each lymphocyte (allelic exclusion) [7,8].

1.3. Junctional Diversity

The joining of V, D, and J gene segments is a very imprecise procedure: the ends of germline segments that are being joined are cleaved randomly [3]. Moreover, the enzyme terminal deoxynucleotidyl transferase randomly adds so called “non-templated nucleotides” (N-bases) at the junctions between the gene segments that are joined together.
These sequence-altering processes further and vastly increase the diversity of antigen receptors and their ability to recognize virtually all possible antigens.

1.4. Affinity Maturation

Naïve B-cells express unmutated IG genes, but after the recognition of antigen by B-lymphocytes in secondary lymphoid organs (lymph nodes), an enzyme called activation-induced cytidine deaminase introduces somatic hypermutations around the productively rearranged V(D)J junction and cells with higher affinity for the antigen are favoured [9,10,11]. This event, called affinity maturation, enables the production of immune receptors with very high affinity to the certain antigen [12].

1.5. Rearrangement Process during Lymphocyte Development

The regulation of the V(D)J recombination in B- and T-cells is accomplished by different accessibility of IG and TR gene due to cell-type specific chromatin structures [13,14]. The IGH D-J joining starts in the ‘common lymphoid progenitor’ stage and IGH V-D-J, IGK and IGL rearrangements are initiated in the pro-B cell compartment. Rearrangement of the IGK locus either leads to IgH/κ expression or is followed by IGK deletion and IGL rearrangement, potentially leading to IgH/λ expression. The successful assembly of IG genes plays a crucial role in guiding B-cell development: B-cells lacking the capacity to rearrange their IG genes are arrested in pro-B cell stage, but the introduction of a rearranged IgH transgene allows the cells to progress to a pre-B cell stage [15,16,17,18,19,20,21].
TR loci rearrange in a highly ordered way. D-D and V-D rearrangements of the TRD locus begin in a pro-T (DN1) stage, followed by D-J rearrangements of the TRD and V-J rearrangements of the TRG locus in pre/pro-T (DN2) stage [22,23]. This is either followed by TRγδ expression or by TRB rearrangement in the pre-T (DN3) stage (D-J rearrangement). The TRB rearrangement is completed in a pre-T (DN4) stage (V-D-J rearrangements) [24]. Lastly, TRA locus rearranges in a double-positive stage [23,25], which is potentially followed by TRαβ expression. Because the TRD locus is nested in the TRA locus (between TRAV and TRAJ gene segments), rearrangement of the TRA locus leads to the deletion of TRD genes. This mechanism ensures that a single cell can either express a TRαβ or TRγδ, but not both.

2. IG/TR Rearrangements in Leukemia

2.1. Leukemic Clones & Oligoclonality

The entire set of antigen receptors with different antigen specificities in one individual is called the immune repertoire. In humans it is believed to be 1011–1012 or higher [1,26]. Thanks to the above-described recombination process, each newly developed B- or T-lymphocyte carries a uniquely rearranged junctional region sequence coding its antigen receptors.
Lymphoid malignancies are clonal diseases. It is therefore commonly believed that all their cells are descendants of a single malignantly transformed B- or T-lymphocyte and that the entire malignant clone carries identical IG/TR V(D)J rearrangement(s). Consequently, the junctional region is considered as a ‘DNA fingerprint’ of each particular clone [27].
Despite the generally accepted monoclonal origin of acute lymphoblastic leukemia (ALL), already early studies using PCR and Southern blotting reported that up to 40% of the B-cell precursor ALL cases are oligoclonal at diagnosis with up to 9 leukemic rearrangements per patient [28,29,30,31,32]. More recently, highly sensitive modern techniques employing next-generation IG/TR sequencing provided evidence that the percentage of patients with oligoclonal IG/TR profiles and also the degree of oligoclonality might be considerably higher [33,34]. Interestingly, oligoclonality at diagnosis is present in 27% of T-ALL patients harboring a cross-lineage IGH rearrangement [35], but TR oligoclonality in T-ALL is rather rare [27].
An oligoclonal IG/TR rearrangements profile is a consequence of continuing rearrangement and secondary rearrangement processes via the active recombinase machinery in these immature lymphoid malignancies [27]. Besides, up to a quarter of CLL patients harbor multiple dominant productive IGH rearrangements [36,37]. Only one third of these cases exhibit two clonal populations with distinct immunophenotypes [38], but each productive IGH rearrangement corresponds to a different B-cell clone also in immunophenotypically monoclonal cases [39].

2.2. IG/TR Rearrangement Profiles in Leukemias

Leukemic transformation leads to a cell differentiation arrest, which has direct impact on the IG/TR gene rearrangement configuration. Therefore, rearrangements in leukemic cells differ from the physiological repertoire and distinct rearrangement profiles can be identified according to age at diagnosis and genetic aberrations [40,41,42].
B-cell precursor ALL results from a leukemic transformation of a lymphoid precursor at an early stage of B-cell differentiation and it is therefore not surprising that over 80% of both adult and pediatric cases carry an IGH rearrangement and around 40% of them carry an IGK rearrangement [43,44]. As might be expected following the same logic, over 90% of T-ALL cases harbor a TRB rearrangement [45], over 80% a TRG and almost 70% (adults) or 40% (children) a TRD rearrangement [43,44]. Exceptionally, however, there are also ALL cases with all IG/TR loci in germline configuration—those are most probably derived from very immature progenitor cells.
Although cross-lineage rearrangements have not been detected in human thymocytes and their frequency in B-cells is very low (<0.5%) [46,47], these so-called illegitimate rearrangements have been identified in leukemic cells besides the lineage-consistent rearrangements: IGH rearrangements in 22% of T-ALL cases [35] and TR rearrangements in 80–90% of patients with B-ALL [48,49]. Cross-lineage rearrangements in B-ALL have several special characteristics, compared to regular rearrangements: TRB rearrangements contain particularly the most downstream Vb gene segments and solely the Jb2 segment, TRG rearrangements involve Jg1 segments in 70% of cases, and 80% of TRD rearrangements are represented by incomplete Vd2-Dd3 or Dd2-Dd3 junctions, which are rare in T-cells [44,48,50,51,52]. TRB rearrangements are virtually absent in pro-B-ALL and in infants, and patients with complete TRB gene rearrangements show a more mature IG/TR profile (higher frequency of IGK, TRG, and Vd2–Ja rearrangements) [53]. Remarkably, the frequency of cross-lineage Vd2-Dd3 rearrangements significantly decreases with age at diagnosis, while cross-lineage TRG rearrangements are rarely found in patients below 2 years of age [27,41]. In T-ALL, the cross-lineage IGH rearrangements are rather immature, as they are characterized by a high frequency of incomplete D-J rearrangements and frequent usage of most downstream Dh6-19 and Dh7-27 and most upstream Jh1 and Jh2 gene segments [35]. Cross-lineage rearrangements are rare in mature B- and T-cell malignancies, probably due to the absence of recombinase activity [54,55,56]. This corresponds with the reported decreasing incidence of cross-lineage TRG rearrangements in more mature B-ALLs: pro-B (57%), common (47%), pre-B (42%), and mature-B (0%) ALL [49]. Also, in more mature T-ALLs with biallelic TRD deletions and completed TRA rearrangements the IGH gene rearrangements are virtually absent [35]. In contrast to CLL, a mature B-cell malignancy, high incidence of non-coding/out-of-frame rearrangements was observed in ALL, suggesting that antigen selection pressure does not play a crucial role in ALL [57].
In CLL, so called stereotyped B-cell receptors are a common phenomenon. Their complementarity-determining region 3 (CDR3) sequences are closely similar (share structural features like V-gene, length, amino acid composition) among unrelated cases, suggesting that stimulation by (auto)antigens may play a role in CLL pathogenesis [58,59]. The IGH/IGK/IGL repertoires in CLL are biased and differ from repertoires in normal B-cells [60,61,62,63]. Additionally, certain V-segments (IGHV3-21 and IGHV1-69) are associated with poor outcome [57]. Furthermore, presence of somatic mutations in variable heavy chain genes defines two CLL subtypes associated with a different clinical course. About half of CLL cases have more than 98% identity to the closest germline V-gene (“unmutated”), which corresponds to inferior outcome compared to patients with “mutated” CLL (less than 98% identity) [64,65].

2.3. Stability and Sensitivity of IG/TR Rearrangements as MRD Targets

Since IG/TR rearrangements are not directly related to the oncogenic process, they may vanish over time due to the outgrowth of subclones or ongoing and secondary rearrangements in leukemic blasts with active IG/TR recombination machinery. This might lead to an underestimation of MRD level if a rearrangement is only present in a small subclone, or even a false-negative MRD result if the rearrangement is fully lost during the disease course. It has therefore been recommended to use at least two leukemia-specific rearrangements to detect MRD in ALL to lower the risk of obtaining a false negative result [66].
Studies comparing the rearrangement profiles at diagnosis and during the disease course or at relapse are almost exclusively focusing on pediatric patients. It has been shown that oligoclonality at diagnosis is the most powerful predictor of ongoing clonal evolution in ALL: particularly in childhood BCP-ALL, significant differences in stability were observed between monoclonal and oligoclonal rearrangements: 89% of monoclonal vs. 40% of oligoclonal rearrangements are preserved at relapse [67]. In this study, roughly 85% of monoclonal IGH and TRD rearrangements remained stable between diagnosis and relapse. Among monoclonal IGK-Kde rearrangements the percentage of stable targets is even higher (95%), probably due to their end-stage character [68]. A study comparing IG/TR profiles at diagnosis and relapse of B-ALL employing high throughput IG/TR sequencing confirmed that the overall stability of IG/TR rearrangements is rather low in (27% of clonal rearrangements were preserved), but also showed that the stability of large clones is way higher (84%) [69]. At relapse, the general characteristics of the IG/TR gene profiles are comparable to those at diagnosis but exhibit a lower degree of oligoclonality and more frequent TRD gene deletions, which fits with the hypothesis of ongoing clonal selection and continuing rearrangements [67]. In T-ALL, the IG/TR rearrangements profiles at diagnosis and relapse are more stable: 97% and 86% of TR rearrangements are preserved at relapse in adult and childhood T-ALL, respectively [70]. TRD rearrangements are the most stable ones (100% of rearrangements preserved at relapse), followed by TRG (89%) and TRB rearrangements (82%) [70].
Besides different stability during the disease course, IG/TR MRD targets also vary in sensitivity of the derived real-time quantitative polymerase chain reaction (RQ-PCR) assays. The sensitivity is primarily determined by the combinatorial and junctional diversity of the CDR3 regions. Therefore, RQ-PCR assays based on rearrangements from IG/TR loci that contain more V/D/J gene segments in their germline sequence (higher combinatorial diversity) generally have higher sensitivity. Similarly, complete rearrangements that contain D-segments (IGH, TRB, TRD V-D-J rearrangements) provide higher sensitivity than complete rearrangements without a D-segment (TRG, IGK rearrangements) and incomplete rearrangements. For example, complete IGH rearrangements represent the most sensitive group of targets, usually reaching the sensitivity of 10−4 [43]. Also, complete TRB rearrangements provide decent sensitivity thanks to their extensive junctional regions [70]. The lower combinatorial diversity in incomplete TRB rearrangements provides an explanation for slightly lower sensitivities in this group of targets [53]. In contrast, the sensitivity of TRG targets is usually considerably limited (a sensitivity of at least 10−4 is reached in less than half of the patients), owing to the restricted size of their junctional regions and the non-specific amplification of highly abundant polyclonal TRG rearrangements in normal T-cells [71]. Intriguingly, TRG rearrangements contain significantly higher number of inserted nucleotides and lower number of deleted nucleotides in T-ALL than in BCP-ALL, which seems to be the most important predictor for reaching good sensitivity [71].

3. Minimal Residual Disease

3.1. MRD as an Important Prognostic Factor in Lymphoid Malignancies

Besides conventional patient-associated (e.g., sex, age at diagnosis, ethnicity) and disease-associated (e.g., white blood cell count at diagnosis, immunophenotype, genetic aberrations) prognostic factors, MRD after induction/consolidation therapy reflecting the treatment efficiency has proven to be most important in predicting patient outcome and risk of relapse in ALL [72,73,74,75,76,77,78,79,80,81,82,83]. The significance of MRD for prognostication has increased over the last decades also in CLL and multiple myeloma. MRD status after the induction phase of treatment is highly significant for predicting (relapse-free) survival, according to several large trials [84,85,86,87,88,89,90,91,92,93,94].
MRD is therefore widely used to identify high risk ALL patients who benefit from treatment intensification and, on the other hand, patients in whom treatment de-escalation, reducing treatment-related toxicity, is feasible. MRD is also monitored in the vast majority of clinical trials, where reaching MRD negativity is considered an important endpoint for the determination of treatment efficiency.

3.2. MRD Detection Techniques

Both low- and high-throughput methods for molecular detection of MRD in lymphoid malignancies either utilize IG/TR rearrangements or fusion gene transcripts associated with the leukemia. IG/TR rearrangements can be detected in 95% [32] of ALL patients and in virtually all CLL patients. Fusion gene transcripts can be detected only in 40–45% of patients with B-ALL and 15–35% of patients with T-ALL [95].
Current standard for molecular MRD detection is an IG/TR-based RQ-PCR, a low-throughput method which has been extensively optimized and standardized within the EuroMRD (www.euromrd.org, accessed on 26 June 2021) Consortium. The sensitivity routinely reaches 10−4 to 10−5 (1 leukemic cell in 10,000 to 100,000 healthy cells). Because each patient’s leukemia-specific IG/TR rearrangements are different, the assay characteristics (sensitivity and the quantitative range) differ among patients and even within a single patient. Therefore, the sensitivity and the quantitative range of IG/TR-based RQ-PCR has to be assessed separately for each individual assay. This is not the case of MRD assays employing fusion gene transcripts since those are shared among patients and no patient-specific assays are therefore necessary. These assays generally reach a sensitivity of 10−5.
Another low-throughput method that can be used for MRD detection is digital droplet PCR (ddPCR). This method builds on the workflow of RQ-PCR. However, thanks to compartmentalization of the reaction into oil droplets and the resulting digital readout, absolute quantification of target DNA without the need of a reference standard curve is possible.
The laboriousness and tardiness associated with the necessity of using patient-specific assays if IG/TR rearrangements are used as markers, can be overcome by next-generation sequencing (NGS). Unlike Sanger sequencing, which provides one representative sequence for all molecules in the PCR, the high throughput of next-generation sequencers allows for obtaining the sequence information for each molecule separately. Using this technique, the same primer set(s) can be used for all patients.
Residual leukemic cells can also be detected by multiparameter flow cytometry (MFC) which can identify cells with leukemia-associated immunophenotype (LAIP), differing from that of normal cells. This approach is way faster compared with the above-mentioned molecular methods and is applicable in >98% of patients with ALL and all patients with CLL [96,97]. The sensitivity of flow cytometry can reach the levels obtained by the molecular methods, provided that a sufficient number of cells are evaluated [97].

3.3. IG/TR-Based MRD Detection by Low and High Throughput Molecular Methods

Regardless of the molecular method used for IG/TR-based MRD detection, characterization of IG/TR rearrangement profile and identification of leukemia-associated IG/TR rearrangement(s) must be performed in each diagnostic sample with a panel of screening PCRs followed by Sanger sequencing or NGS of clonal PCR products (Figure 1). Once the sequence of the leukemia-associated rearrangement(s) is acquired, it can be used as a marker for the detection of MRD.
Until recently, multiplex assays developed in a European BIOMED-2 collaborative study [98] and low-throughput Sanger sequencing have been commonly used by most laboratories. However, with the advent of NGS, new assays for amplicon NGS have been developed by the EuroClonality-NGS working group (https://euroclonalityngs.org/, accessed on 26 July 2021) [99], which is formed by several European groups with expertise in assay design, immunobiology, bioinformatics and clinical application of IG/TR analysis in hemato-oncology, pathology, and immunology. Improved primer sets for use with NGS were designed for a better coverage and equal amplification efficiency of all rearrangements. NGS-based approaches perform better in samples containing bi-allelic rearrangements, polyclonal background and in oligoclonal samples, where the low-throughput sequencing provides mixed sequences that cannot be used for patient-specific primer design [99] (Figure 2). The NGS-based marker identification has already been introduced in many MRD laboratories and the EuroMRD Consortium has already included NGS-based marker identification in its quality control rounds.
Despite current advances in the field of molecular MRD detection, RQ-PCR of clonal IG/TR gene rearrangements remains the most widely used molecular method for MRD assessment due to the lack of standardization of the modern methods. It requires junctional region-specific forward primer, reverse primer or probe (called allele-specific oligonucleotides (ASO)) designed based on the leukemia-associated sequence obtained during the initial marker screening. The most common strategy is the ‘ASO forward primer approach’, using germline probes and reverse primers located in the J gene segments. It has several advantages compared to the ‘ASO reverse primer’ and ‘ASO probe’ approaches: (1) because the number of J gene segments is lower than the number of V gene segments, lower number of germline probes and reverse primers is needed; (2) the binding sites of germline probes and primers located in the J-gene segments remain unaffected in cases with ongoing rearrangements, V replacements and somatic hypermutations. The sensitivity and quantitative range of each assay must be determined separately based on the performance of a dilution curve from the diagnostic material of each particular patient. These must be taken into account for the final interpretation of RQ-PCR MRD results according to the international guidelines [100]. The need of a dilution curve can limit the applicability of the RQ-PCR in patients with insufficient amount of diagnostic DNA.
The patient-specific primers/probes are also indispensable for ddPCR-based MRD detection, therefore even in this setting, the laborious design of patient-specific assays cannot be avoided. ddPCR can however overcome certain limitations of IG/TR-based RQ-PCR and can therefore serve as an attractive low-throughput alternative for MRD detection. In particular, ddPCR provides an absolute MRD quantification without the need of a dilution curve. This is crucial for high-risk patients in whom repeated MRD monitoring is required, because the amount of diagnostic material necessary for the standard curve is limited. Still, no guidelines for ddPCR-based MRD detection have been defined so far and its prognostic power is still under investigation. The EuroMRD Consortium is currently putting great effort into standardization of the method for MRD detection in lymphoid malignancies.
On the contrary, no patient-specific oligonucleotides are needed for NGS-based MRD detection. The same multiplex primer set used for marker identification can be used for MRD detection in all patients, hereby avoiding the cumbersome design and testing of patient-specific ASO RQ-PCR assays. Although the wet-lab part of NGS-based MRD detection is more straightforward when compared to low throughput methods, the subsequent data analysis is challenging and requires expertise and specialized bioinformatics tools. As already mentioned above, NGS provides a sequence of each single IG/TR rearrangement in the tested sample. This is useful for detection of emerging leukemic (sub)clones resulting from the ongoing rearrangement activity and also assessing the background IG/TR repertoire. Nevertheless, as with the ddPCR, the standardization of the method and assessment of its prognostic power on large patient cohorts is still in progress within the EuroClonality-NGS working group.
Although NGS and ddPCR have not yet been fully implemented into routine practice, there are hints that both methods possess appreciable advantages over RQ-PCR. Particularly samples which are positive below the quantitative range (pos < QR) of RQ-PCR can often be assessed more confidently: thanks to the more specific readout of NGS, false-positive RQ-PCR results which might occur due to the nonspecific amplification can be reliably distinguished from truly positive samples [101]; ddPCR is able to provide an accurate MRD quantification in 20–45% of pos < QR samples thanks to its absolute quantification without the need of a standard curve [102,103,104,105]. Another frequently mentioned advantage of the modern methods is the improved sensitivity. However, such conclusions must be interpreted cautiously. The sensitivity, which is often demonstrated on artificially prepared MRD samples (serial dilutions of patient material/cell lines/plasmids in healthy mononuclear cells), may not necessarily reflect the reality in the clinical setting. Fundamental factors influencing the sensitivity are the number of analyzed cells (DNA amount) and the sequencing coverage. While increasing the sequencing coverage is only a matter of costs, the amount of valuable patient DNA is an inevitable limiting factor. One might assume that to reach a sensitivity of 1 in 1 million (10−6), it is sufficient to analyze DNA from a million cells (equivalent to roughly 6.5 μg). Nevertheless, several critical factors increase the requirements on the cell quantity significantly. Firstly, randomness of subsampling must be taken into account. According to the Poisson statistics, threefold amount of DNA (i.e., 20 μg for the sensitivity of 10−6) is necessary to confidently detect a single cell in 95% of cases [106]. Additionally, loss of material during numerous laboratory steps and an unequal amplification efficiency for different IG/TR rearrangements further increase the DNA quantity requirements.
Figure 2. Normalized abundances of IG/TR clones in the diagnostic sample of a patient with B-cell precursor ALL. The sample was analysed using the EuroClonality-NGS assays [99]. Clone abundances were normalized using the EuroClonality-NGS spike-in controls [107]. Clones with an abundance <0.01% are not shown. Leukemia-associated clones which can be used for MRD-detection are shown as red dots, background physiological rearrangements as blue dots.
Figure 2. Normalized abundances of IG/TR clones in the diagnostic sample of a patient with B-cell precursor ALL. The sample was analysed using the EuroClonality-NGS assays [99]. Clone abundances were normalized using the EuroClonality-NGS spike-in controls [107]. Clones with an abundance <0.01% are not shown. Leukemia-associated clones which can be used for MRD-detection are shown as red dots, background physiological rearrangements as blue dots.
Genes 12 00979 g002

4. Summary

Thanks to their uniqueness, IG/TR rearrangements can be used as strongly specific molecular markers of lymphocyte clones. Often being referred to as ‘molecular fingerprints’, virtually any physiological or malignant lymphocyte clone can be detected using IG/TR rearrangements as markers. Although modern molecular methods bring certain benefits, mainly in the form of higher specificity and possibly also additional information on the background clones, their standardization and validation in a clinical setting has not yet been finalized. Nonetheless, the modern methods are already being very useful in a research setting and in special cases where RQ-PCR does not provide unambiguous results. Whether any of the aforementioned molecular methods is used, the interpretation of the IG/TR data is a highly complex topic, requiring a high level of expertise and should therefore be only performed by well-trained professionals.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

M.B. received personal fees from Incyte (advisory board) and Roche Pharma AG, financial support for reference diagnostics from Affimed and Regeneron, grants and personal fees from Amgen (advisory board, speakers bureau, travel support), and personal fees from Janssen (speakers bureau), all outside the submitted work. The remaining authors declare no competing financial interests.

References

  1. Murphy, K. Janeway’s Immunobiology, 8th ed.; Garland Science, Taylor & Francis Group LLC: New York, NY, USA, 2012; ISBN 978-0-8153-4243-4. [Google Scholar]
  2. Jung, D.; Giallourakis, C.; Mostoslavsky, R.; Alt, F.W. Mechanism and control of V(D)J recombination at the immunoglobulin heavy chain locus. Annu. Rev. Immunol. 2006, 24, 541–570. [Google Scholar] [CrossRef] [Green Version]
  3. Tonegawa, S. Somatic generation of antibody diversity. Nature 1983, 302, 575–581. [Google Scholar] [CrossRef]
  4. Van Dongen, J.J.M.; Comans-Bitter, W.M.; Wolvers-Tettero, I.L.M.; Borst, J. Development of human T lymphocytes and their thymus-dependency. Thymus 1990, 16, 207–234. [Google Scholar] [PubMed]
  5. Davis, M.M.; Bjorkman, P.J. T-cell antigen receptor genes and T-cell recognition. Nature 1988, 334, 395–402. [Google Scholar] [CrossRef] [PubMed]
  6. Szczepański, T.; Van Der Velden, V.H.J.; Hoogeveen, P.G.; De Bie, M.; Jacobs, D.C.H.; Van Wering, E.R.; Van Dongen, J.J.M. Vδ2-Jα rearrangements are frequent in precursor-B-acute lymphoblastic leukemia but rare in normal lymphoid cells. Blood 2004, 103, 3798–3804. [Google Scholar] [CrossRef]
  7. Fröland, S.S.; Natvig, J.B. Class, subclass, and allelic exclusion of membrane-bound Ig of human B lymphocytes. J. Exp. Med. 1972, 136, 409–414. [Google Scholar] [CrossRef]
  8. Aifantis, I.; Buer, J.; Von Boehmer, H.; Azogui, O. Essential Role of the Pre-T Cell Receptor in Allelic Exclusion of the T Cell Receptor β locus. Immunity 1997, 7, 601–607. [Google Scholar] [CrossRef] [Green Version]
  9. Di Noia, J.M.; Neuberger, M.S. Molecular Mechanisms of Antibody Somatic Hypermutation. Annu. Rev. Biochem. 2007, 76, 1–22. [Google Scholar] [CrossRef]
  10. Tomlinson, I.M.; Walter, G.; Jones, P.T.; Dear, P.H.; Sonnhammer, E.L.; Winter, G. The imprint of somatic hypermutation on the repertoire of human germline V genes. J. Mol. Biol. 1996, 256, 813–817. [Google Scholar] [CrossRef] [Green Version]
  11. Neuberger, M.S.; Milstein, C. Somatic hypermutation. Curr. Opin. Immunol. 1995, 7, 248–254. [Google Scholar] [CrossRef]
  12. Bye, J.M.; Carter, C.; Cui, Y.; Gorick, B.D.; Songsivilai, S.; Winter, G.; Hughes-Jones, N.C.; Marks, J.D. Germline variable region gene segment derivation of human monoclonal anti-Rh(D) antibodies: Evidence for affinity maturation by somatic hypermutation and repertoire shift. J. Clin. Investig. 1992, 90, 2481–2490. [Google Scholar] [CrossRef] [Green Version]
  13. Kosak, S.T.; Skok, J.A.; Medina, K.L.; Riblet, R.; Le Beau, M.M.; Fisher, A.G.; Singh, H. Subnuclear compartmentalization of immunoglobulin loci during lymphocyte development. Science 2002, 296, 158–162. [Google Scholar] [CrossRef]
  14. Stanhope-Baker, P.; Hudson, K.M.; Shaffer, A.L.; Constantinescu, A.; Schlissel, M.S. Cell Type-Specific Chromatin Structure Determines the Targeting of V(D)J Recombinase Activity In Vitro. Cell 1996, 85, 887–897. [Google Scholar] [CrossRef] [Green Version]
  15. Miyazaki, K.; Miyazaki, M.; Murre, C. The establishment of B versus T cell identity. Trends Immunol. 2014, 35, 205–210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Van Zelm, M.C.; van der Burg, M.; de Ridder, D.; Barendregt, B.H.; de Haas, E.F.E.; Reinders, M.J.T.; Lankester, A.C.; Révész, T.; Staal, F.J.T.; van Dongen, J.J.M. Ig Gene Rearrangement Steps Are Initiated in Early Human Precursor B Cell Subsets and Correlate with Specific Transcription Factor Expression. J. Immunol. 2005, 175, 5912–5922. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Constantinescu, A.; Schlissel, M.S. Changes in locus-specific V(D)J recombinase activity induced by immunoglobulin gene products during B cell development. J. Exp. Med. 1997, 185, 609–620. [Google Scholar] [CrossRef] [Green Version]
  18. Mombaerts, P.; Iacomini, J.; Johnson, R.S.; Herrup, K.; Tonegawa, S.; Papaioannou, V.E. RAG-1-deficient mice have no mature B and T lymphocytes. Cell 1992, 68, 869–877. [Google Scholar] [CrossRef]
  19. Shinkai, Y.; Rathbun, G.; Lam, K.P.; Oltz, E.M.; Stewart, V.; Mendelsohn, M.; Charron, J.; Datta, M.; Young, F.; Stall, A.M.; et al. RAG-2-deficient mice lack mature lymphocytes owing to inability to initiate V(D)J rearrangement. Cell 1992, 68, 855–867. [Google Scholar] [CrossRef]
  20. Spanopoulou, E.; Roman, C.A.J.; Corcoran, L.M.; Schlissel, M.S.; Silver, D.P.; Nemazee, D.; Nussenzweig, M.C.; Shinton, S.A.; Hardy, R.R.; Baltimore, D. Functional immunoglobulin transgenes guide ordered B-cell differentiation in Rag-1-deficient mice. Genes Dev. 1994, 8, 1030–1042. [Google Scholar] [CrossRef] [Green Version]
  21. Young, F.; Ardman, B.; Shinkai, Y.; Lansford, R.; Blackwell, T.K.; Mendelsohn, M.; Rolink, A.; Melchers, F.; Alt, F.W. Influence of immunoglobulin heavy- and light-chain expression on B-cell differentiation. Genes Dev. 1994, 8, 1043–1057. [Google Scholar] [CrossRef] [Green Version]
  22. Capone, M.; Hockett, R.D.; Zlotnik, A. Kinetics of T cell receptor β, γ, and δ rearrangements during adult thymic development: T cell receptor rearrangements are present in CD44+CD25+ Pro-T thymocytes. Proc. Natl. Acad. Sci. USA 1998, 95, 12522–12527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Staal, F.J.T.; Weerkamp, F.; Langerak, A.W.; Hendriks, R.W.; Clevers, H.C. Transcriptional Control of T Lymphocyte Differentiation. Stem Cells 2001, 19, 165–179. [Google Scholar] [CrossRef] [Green Version]
  24. Von Boehmer, H. Selection of the T-cell repertoire: Receptor-controlled checkpoints in T-cell development. Adv. Immunol. 2004, 84, 201–238. [Google Scholar] [CrossRef]
  25. Dik, W.A.; Pike-Overzet, K.; Weerkamp, F.; De Ridder, D.; De Haas, E.F.E.; Baert, M.R.M.; Van Der Spek, P.; Koster, E.E.L.; Reinders, M.J.T.; Van Dongen, J.J.M.; et al. New insights on human T cell development by quantitative T cell receptor gene rearrangement studies and gene expression profiling. J. Exp. Med. 2005, 201, 1715–1723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Imkeller, K.; Wardemann, H. Assessing human B cell repertoire diversity and convergence. Immunol. Rev. 2018, 284, 51–66. [Google Scholar] [CrossRef]
  27. Szczepański, T.; Flohr, T.; Van Der Velden, V.H.J.; Bartram, C.R.; Van Dongen, J.J.M. Molecular monitoring of residual disease using antigen receptor genes in childhood acute lymphoblastic leukaemia. Best Pract. Res. Clin. Haematol. 2002, 15, 37–57. [Google Scholar] [CrossRef] [PubMed]
  28. Beishuizen, A.; Verhoeven, M.A.; van Wering, E.R.; Hählen, K.; Hooijkaas, H.; van Dongen, J.J. Analysis of Ig and T-cell receptor genes in 40 childhood acute lymphoblastic leukemias at diagnosis and subsequent relapse: Implications for the detection of minimal residual disease by polymerase chain reaction analysis. Blood 1994, 83, 2238–2247. [Google Scholar] [CrossRef] [Green Version]
  29. Beishuizen, A.; Hählen, K.; Hagemeijer, A.; Verhoeven, M.A.; Hooijkaas, H.; Adriaansen, H.J.; Wolvers-Tettero, I.L.; van Wering, E.R.; van Dongen, J.J. Multiple rearranged immunoglobulin genes in childhood acute lymphoblastic leukemia of precursor B-cell origin. Leukemia 1991, 5, 657–667. [Google Scholar] [PubMed]
  30. Steenbergen, E.J.; Verhagen, O.J.; van Leeuwen, E.F.; von dem Borne, A.E.; van der Schoot, C.E. Distinct ongoing Ig heavy chain rearrangement processes in childhood B-precursor acute lymphoblastic leukemia. Blood 1993, 82, 581–589. [Google Scholar] [CrossRef] [Green Version]
  31. Kitchingman, G.R. Immunoglobulin heavy chain gene VH-D junctional diversity at diagnosis in patients with acute lymphoblastic leukemia. Blood 1993, 81, 775–782. [Google Scholar] [CrossRef] [Green Version]
  32. Van Der Velden, V.H.J.; Van Dongen, J.J.M. MRD detection in acute lymphoblastic leukemia patients using Ig/TCR gene rearrangements as targets for real-time quantitative PCR. Methods Mol. Biol. 2009, 538, 115–150. [Google Scholar] [CrossRef] [PubMed]
  33. Theunissen, P.M.J.; van Zessen, D.; Stubbs, A.P.; Faham, M.; Zwaan, C.M.; van Dongen, J.J.M.; Van Der Velden, V.H.J. Antigen receptor sequencing of paired bone marrow samples shows homogeneous distribution of acute lymphoblastic leukemia subclones. Haematologica 2017, 102, 1869–1877. [Google Scholar] [CrossRef] [Green Version]
  34. Gawad, C.; Pepin, F.; Carlton, V.E.H.; Klinger, M.; Logan, A.C.; Miklos, D.B.; Faham, M.; Dahl, G.; Lacayo, N. Massive evolution of the immunoglobulin heavy chain locus in children with B precursor acute lymphoblastic leukemia. Blood 2012, 120, 4407–4417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Szczepański, T.; Pongers-Willemse, M.J.; Langerak, A.W.; Harts, W.A.; Wijkhuijs, A.J.M.; van Wering, E.R.; van Dongen, J.J.M. Ig Heavy Chain Gene Rearrangements in T-Cell Acute Lymphoblastic Leukemia Exhibit Predominant Dh6-19 and Dh7-27 Gene Usage, Can Result in Complete V-D-J Rearrangements, and Are Rare in T-Cell Receptor αβ Lineage. Blood 1999, 93, 4079–4085. [Google Scholar] [CrossRef]
  36. Stamatopoulos, B.; Timbs, A.; Bruce, D.; Smith, T.; Clifford, R.; Robbe, P.; Burns, A.; Vavoulis, D.V.; Lopez, L.; Antoniou, P.; et al. Targeted deep sequencing reveals clinically relevant subclonal IgHV rearrangements in chronic lymphocytic leukemia. Leukemia 2017, 31, 837–845. [Google Scholar] [CrossRef]
  37. Langerak, A.W.; Davi, F.; Ghia, P.; Hadzidimitriou, A.; Murray, F.; Potter, K.N.; Rosenquist, R.; Stamatopoulos, K.; Belessi, C. Immunoglobulin sequence analysis and prognostication in CLL: Guidelines from the ERIC review board for reliable interpretation of problematic cases. Leukemia 2011, 25, 979–984. [Google Scholar] [CrossRef] [Green Version]
  38. Plevova, K.; Francova, H.S.; Burckova, K.; Brychtova, Y.; Doubek, M.; Pavlova, S.; Malcikova, J.; Mayer, J.; Tichy, B.; Pospisilova, S. Multiple productive immunoglobulin heavy chain gene rearrangements in chronic lymphocytic leukemia are mostly derived from independent clones. Haematologica 2014, 99, 329–338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Brazdilova, K.; Plevova, K.; Skuhrova Francova, H.; Kockova, H.; Borsky, M.; Bikos, V.; Malcikova, J.; Oltova, A.; Kotaskova, J.; Tichy, B.; et al. Multiple productive IGH rearrangements denote oligoclonality even in immunophenotypically monoclonal CLL. Leukemia 2018, 32, 234–236. [Google Scholar] [CrossRef]
  40. Hübner, S.; Cazzaniga, G.; Flohr, T.; van der Velden, V.H.J.; Konrad, M.; Pötschger, U.; Basso, G.; Schrappe, M.; van Dongen, J.J.M.; Bartram, C.R.; et al. High incidence and unique features of antigen receptor gene rearrangements in TEL–AML1-positive leukemias. Leukemia 2004, 18, 84–91. [Google Scholar] [CrossRef] [Green Version]
  41. Brumpt, C.; Delabesse, E.; Beldjord, K.; Davi, F.; Cayuela, J.M.; Millien, C.; Villarese, P.; Quartier, P.; Buzyn, A.; Valensi, F.; et al. The incidence of clonal T-cell receptor rearrangements in B-cell precursor acute lymphoblastic leukemia varies with age and genotype. Blood 2000, 96, 2254–2261. [Google Scholar] [CrossRef]
  42. Van der Velden, V.H.J.; Szczepanski, T.; Wijkhuijs, J.M.; Hart, P.G.; Hoogeveen, P.G.; Hop, W.C.J.; van Wering, E.R.; van Dongen, J.J.M. Age-related patterns of immunoglobulin and T-cell receptor gene rearrangements in precursor-B-ALL: Implications for detection of minimal residual disease. Leukemia 2003, 17, 1834–1844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Flohr, T.; Schrauder, A.; Cazzaniga, G.; Panzer-Grümayer, R.; van der Velden, V.; Fischer, S.; Stanulla, M.; Basso, G.; Niggli, F.K.; Schäfer, B.W.; et al. Minimal residual disease-directed risk stratification using real-time quantitative PCR analysis of immunoglobulin and T-cell receptor gene rearrangements in the international multicenter trial AIEOP-BFM ALL 2000 for childhood acute lymphoblastic leukemia. Leukemia 2008, 22, 771–782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Szczepański, T.; Langerak, A.W.; Wolvers-Tettero, I.L.M.; Ossenkoppele, G.J.; Verhoef, G.; Stul, M.; Petersen, E.J.; De Bruijn, M.A.C.; Van’t Veer, M.B.; Van Dongen, J.J.M. Immunoglobulin and T cell receptor gene rearrangement patterns in acute lymphoblastic leukemia are less mature in adults than in children: Implications for selection of PCR targets for detection of minimal residual disease. Leukemia 1998, 12, 1081–1088. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Brüggemann, M.; van der Velden, V.H.J.; Raff, T.; Droese, J.; Ritgen, M.; Pott, C.; Wijkhuijs, A.J.; Gökbuget, N.; Hoelzer, D.; van Wering, E.R.; et al. Rearranged T-cell receptor beta genes represent powerful targets for quantification of minimal residual disease in childhood and adult T-cell acute lymphoblastic leukemia. Leukemia 2004, 18, 709–719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Greaves, M.F.; Chan, L.C.; Furley, A.J.; Watt, S.M.; Molgaard, H.V. Lineage promiscuity in hemopoietic differentiation and leukemia. Blood 1986, 67, 1–11. [Google Scholar] [CrossRef] [Green Version]
  47. Bertrand, F.E.; Billips, L.G.; Burrows, P.D.; Gartland, G.L.; Kubagawa, H.; Schroeder, H.W. Ig D(H) gene segment transcription and rearrangement before surface expression of the pan-B-cell marker CD19 in normal human bone marrow. Blood 1997, 90, 736–744. [Google Scholar] [CrossRef] [Green Version]
  48. Szczepański, T.; Beishuizen, A.; Pongers-Willemse, M.J.; Hählen, K.; Van Wering, E.R.; Wijkhuijs, A.J.M.; Tibbe, G.J.M.; De Bruijn, M.A.C.; Van Dongen, J.J.M. Cross-lineage T cell receptor gene rearrangements occur in more than ninety percent of childhood precursor-B acute lymphoblastic leukemias: Alternative PCR targets for detection of minimal residual disease. Leukemia 1999, 13, 196–205. [Google Scholar] [CrossRef] [Green Version]
  49. Meleshko, A.N.; Belevtsev, M.V.; Savitskaja, T.V.; Potapnev, M.P. The incidence of T-cell receptor gene rearrangements in childhood B-lineage acute lymphoblastic leukemia is related to immunophenotype and fusion oncogene expression. Leuk. Res. 2006, 30, 795–800. [Google Scholar] [CrossRef]
  50. Biondi, A.; Francia di Celle, P.; Rossi, V.; Casorati, G.; Matullo, G.; Giudici, G.; Foa, R.; Migone, N. High prevalence of T-cell receptor V delta 2-(D)-D delta 3 or D delta 1/2-D delta 3 rearrangements in B-precursor acute lymphoblastic leukemias. Blood 1990, 75, 1834–1840. [Google Scholar] [CrossRef] [Green Version]
  51. Hara, J.; Benedict, S.H.; Champagne, E.; Takihara, Y.; Mak, T.W.; Minden, M.; Gelfand, E.W. T cell receptor δ gene rearrangements in acute lymphoblastic leukemia. J. Clin. Investig. 1988, 82, 1974–1982. [Google Scholar] [CrossRef]
  52. Felix, C.A.; Poplack, D.G. Characterization of acute lymphoblastic leukemia of childhood by immunoglobulin and T-cell receptor gene patterns. Leukemia 1991, 5, 1015–1025. [Google Scholar] [CrossRef] [PubMed]
  53. Van der Velden, V.H.J.; Brüggemann, M.; Hoogeveen, P.G.; de Bie, M.; Hart, P.G.; Raff, T.; Pfeifer, H.; Lüschen, S.; Szczepański, T.; van Wering, E.R.; et al. TCRB gene rearrangements in childhood and adult precursor-B-ALL: Frequency, applicability as MRD-PCR target, and stability between diagnosis and relapse. Leukemia 2004, 18, 1971–1980. [Google Scholar] [CrossRef]
  54. Szczepański, T.; Pongers-Willemse, M.J.; Langerak, A.W.; Van Dongen, J.J.M. Unusual immunoglobulin and T-cell receptor gene rearrangement patterns in acute lymphoblastic leukemias. Curr. Top. Microbiol. Immunol. 1999, 246, 205–215. [Google Scholar] [PubMed]
  55. Foroni, L.; Foldi, J.; Matutes, E.; Catovsky, D.; O’Connor, N.J.O.; Baer, R.; Forster, A.; Rabbitts, T.H.; Luzzatto, L. α, β and γ T-cell receptor genes: Rearrangements correlate with haematological phenotype in T cell leukaemias. Br. J. Haematol. 1987, 67, 307–318. [Google Scholar] [CrossRef] [PubMed]
  56. Kneba, M.; Bergholz, M.; Bolz, I.; Hulpke, M.; Bätge, R.; Schauer, A.; Krieger, G. Heterogeneity of immunoglobulin gene rearrangements in B-cell lymphomas. Int. J. Cancer 1990, 45, 609–613. [Google Scholar] [CrossRef] [PubMed]
  57. Rai, L.; Casanova, A.; Moorman, A.V.; Richards, S.; Buck, G.; Goldstone, A.H.; Fielding, A.K.; Foroni, L. Antigen receptor gene rearrangements reflect on the heterogeneity of adult Acute Lymphoblastic Leukaemia (ALL) with implications of cell-origin of ALL subgroups-A UKALLXII study. Br. J. Haematol. 2010, 148, 394–401. [Google Scholar] [CrossRef]
  58. Stamatopoulos, K.; Belessi, C.; Moreno, C.; Boudjograh, M.; Guida, G.; Smilevska, T.; Belhoul, L.; Stella, S.; Stavroyianni, N.; Crespo, M.; et al. Over 20% of patients with chronic lymphocytic leukemia carry stereotyped receptors: Pathogenetic implications and clinical correlations. Blood 2007, 109, 259–270. [Google Scholar] [CrossRef]
  59. Landgren, O.; Albitar, M.; Ma, W.; Abbasi, F.; Hayes, R.B.; Ghia, P.; Marti, G.E.; Caporaso, N.E. B-Cell Clones as Early Markers for Chronic Lymphocytic Leukemia. N. Engl. J. Med. 2009, 360, 659–667. [Google Scholar] [CrossRef]
  60. Chiorazzi, N.; Ferrarini, M. B cell chronic lymphocytic leukemia: Lessons learned from studies of the B cell antigen receptor. Annu. Rev. Immunol. 2003, 21, 841–894. [Google Scholar] [CrossRef] [Green Version]
  61. Fais, F.; Ghiotto, F.; Hashimoto, S.; Sellars, B.; Valetto, A.; Allen, S.L.; Schulman, P.; Vinciguerra, V.P.; Rai, K.; Rassenti, L.Z.; et al. Chronic lymphocytic leukemia B cells express restricted sets of mutated and unmutated antigen receptors. J. Clin. Investig. 1998, 102, 1515–1525. [Google Scholar] [CrossRef]
  62. Stevenson, F.K.; Caligaris-Cappio, F. Chronic lymphocytic leukemia: Revelations from the B-cell receptor. Blood 2004, 103, 4389–4395. [Google Scholar] [CrossRef] [PubMed]
  63. Abusedra, A.; Joshi, R.; Bybee, A.; Apperley, J.F.; Wagner, S.D. Vλ genes in chronic lymphocytic leukaemia: Highly skewed V gene segment usage with similar CDR3 sequences. Leukemia 2008, 22, 1073–1075. [Google Scholar] [CrossRef] [PubMed]
  64. Hamblin, T.J.; Davis, Z.; Gardiner, A.; Oscier, D.G.; Stevenson, F.K. Unmutated Ig V(H) genes are associated with a more aggressive form of chronic lymphocytic leukemia. Blood 1999, 94, 1848–1854. [Google Scholar] [CrossRef] [PubMed]
  65. Damle, R.N.; Wasil, T.; Fais, F.; Ghiotto, F.; Valetto, A.; Allen, S.L.; Buchbinder, A.; Budman, D.; Dittmar, K.; Kolitz, J.; et al. Ig V Gene Mutation Status and CD38 Expression As Novel Prognostic Indicators in Chronic Lymphocytic Leukemia. Blood 1999, 94, 1840–1847. [Google Scholar] [CrossRef] [PubMed]
  66. Baruchel, A.; Cayuela, J.-M.; Macintyre, E.; Berger, I.R.; Sigaux, F. Assessment of clonal evolution at Ig/TCR loci in acute lymphoblastic leukaemia by single-strand conformation polymorphism studies and highly resolutive PCR derived methods: Implication for a general strategy of minimal residual disease detection. Br. J. Haematol. 1995, 90, 85–93. [Google Scholar] [CrossRef]
  67. Szczeparński, T.; Willemse, M.J.; Brinkhof, B.; Van Wering, E.R.; Van Der Burg, M.; Van Dongen, J.J.M. Comparative analysis of Ig and TCR gene rearrangements at diagnosis and at relapse of childhood precursor-B-ALL provides improved strategies for selection of stable PCR targets for monitoring of minimal residual disease. Blood 2002, 99, 2315–2323. [Google Scholar] [CrossRef] [Green Version]
  68. Van der Velden, V.H.J.; Willemse, M.J.; van der Schoot, C.E.; Hählen, K.; van Wering, E.R.; van Dongen, J.J.M. Immunoglobulin kappa deleting element rearrangements in precursor-B acute lymphoblastic leukemia are stable targets for detection of minimal residual disease by real-time quantitative PCR. Leukemia 2002, 16, 928–936. [Google Scholar] [CrossRef] [Green Version]
  69. Theunissen, P.M.J.; de Bie, M.; van Zessen, D.; de Haas, V.; Stubbs, A.P.; van der Velden, V.H.J. Next-generation antigen receptor sequencing of paired diagnosis and relapse samples of B-cell acute lymphoblastic leukemia: Clonal evolution and implications for minimal residual disease target selection. Leuk. Res. 2019, 76, 98–104. [Google Scholar] [CrossRef]
  70. Szczepański, T.; van der Velden, V.H.J.; Raff, T.; Jacobs, D.C.H.; van Wering, E.R.; Brüggemann, M.; Kneba, M.; van Dongen, J.J.M. Comparative analysis of T-cell receptor gene rearrangements at diagnosis and relapse of T-cell acute lymphoblastic leukemia (T-ALL) shows high stability of clonal markers for monitoring of minimal residual disease and reveals the occurence of second T-ALL. Leukemia 2003, 17, 2149–2156. [Google Scholar] [CrossRef] [Green Version]
  71. Van der Velden, V.H.J.; Wijkhuijs, J.M.; Jacobs, D.C.H.; van Wering, E.R.; van Dongen, J.J.M. T cell receptor gamma gene rearrangements as targets for detection of minimal residual disease in acute lymphoblastic leukemia by real-time quantitative PCR analysis. Leukemia 2002, 16, 1372–1380. [Google Scholar] [CrossRef] [Green Version]
  72. Reiter, A.; Schrappe, M.; Ludwig, W.D.; Hiddemann, W.; Sauter, S.; Henze, G.; Zimmermann, M.; Lampert, F.; Havers, W.; Niethammer, D. Chemotherapy in 998 unselected childhood acute lymphoblastic leukemia patients. Results and conclusions of the multicenter trial ALL-BFM 86. Blood 1994, 84, 3122–3133. [Google Scholar] [CrossRef]
  73. Gökbuget, N.; Kneba, M.; Raff, T.; Trautmann, H.; Bartram, C.R.; Arnold, R.; Fietkau, R.; Freund, M.; Ganser, A.; Ludwig, W.D.; et al. Adult patients with acute lymphoblastic leukemia and molecular failure display a poor prognosis and are candidates for stem cell transplantation and targeted therapies. Blood 2012, 120, 1868–1876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Bassan, R.; Brüggemann, M.; Radcliffe, H.S.; Hartfield, E.; Kreuzbauer, G.; Wetten, S. A systematic literature review and metaanalysis of minimal residual disease as a prognostic indicator in adult B-cell acute lymphoblastic leukemia. Haematologica 2019, 104, 2028–2039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Berry, D.A.; Zhou, S.; Higley, H.; Mukundan, L.; Fu, S.; Reaman, G.H.; Wood, B.L.; Kelloff, G.J.; Jessup, J.M.; Radich, J.P. Association of minimal residual disease with clinical outcome in pediatric and adult acute lymphoblastic leukemia: A meta-analysis. JAMA Oncol. 2017, 3, e170580. [Google Scholar] [CrossRef]
  76. Beldjord, K.; Chevret, S.; Asnafi, V.; Huguet, F.; Boulland, M.L.; Leguay, T.; Thomas, X.; Cayuela, J.M.; Grardel, N.; Chalandon, Y.; et al. Oncogenetics and minimal residual disease are independent outcome predictors in adult patients with acute lymphoblastic leukemia. Blood 2014, 123, 3739–3749. [Google Scholar] [CrossRef] [PubMed]
  77. Bassan, R.; Spinelli, O.; Oldani, E.; Intermesoli, T.; Tosi, M.; Peruta, B.; Rossi, G.; Borlenghi, E.; Pogliani, E.M.; Terruzzi, E.; et al. Improved risk classification for risk-specific therapy based on the molecular study of minimal residual disease (MRD) in adult acute lymphoblastic leukemia (ALL). Blood 2009, 113, 4153–4162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Ribera, J.M.; Oriol, A.; Morgades, M.; Montesinos, P.; Sarrà, J.; González-Campos, J.; Brunet, S.; Tormo, M.; Fernández-Abellán, P.; Guàrdia, R.; et al. Treatment of high-risk Philadelphia chromosome-negative acute lymphoblastic leukemia in adolescents and adults according to early cytologic response and minimal residual disease after consolidation assessed by flow cytometry: Final results of the PETHEMA ALL-AR-03 trial. J. Clin. Oncol. 2014, 32, 1595–1604. [Google Scholar] [CrossRef] [PubMed]
  79. Van der Velden, V.H.J.; Corral, L.; Valsecchi, M.G.; Jansen, M.W.J.C.; De Lorenzo, P.; Cazzaniga, G.; Panzer-Grümayer, E.R.; Schrappe, M.; Schrauder, A.; Meyer, C.; et al. Prognostic significance of minimal residual disease in infants with acute lymphoblastic leukemia treated within the Interfant-99 protocol. Leukemia 2009, 23, 1073–1079. [Google Scholar] [CrossRef]
  80. Fronkova, E.; Mejstrikova, E.; Avigad, S.; Chik, K.W.; Castillo, L.; Manor, S.; Reznickova, L.; Valova, T.; Zdrahalova, K.; Hrusak, O.; et al. Minimal residual disease (MRD) analysis in the non-MRD-based ALL IC-BFM 2002 protocol for childhood ALL: Is it possible to avoid MRD testing? Leukemia 2008, 22, 989–997. [Google Scholar] [CrossRef]
  81. Schultz, K.R.; Pullen, D.J.; Sather, H.N.; Shuster, J.J.; Devidas, M.; Borowitz, M.J.; Carroll, A.J.; Heerema, N.A.; Rubnitz, J.E.; Loh, M.L.; et al. Risk- and response-based classification of childhood B-precursor acute lymphoblastic leukemia: A combined analysis of prognostic markers from the Pediatric Oncology Group (POG) and Children’s Cancer Group (CCG). Blood 2007, 109, 926–935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Van Dongen, J.J.M.; Seriu, T.; Panzer-Grümayer, E.R.; Biondi, A.; Pongers-Willemse, M.J.; Corral, L.; Stolz, F.; Schrappe, M.; Masera, G.; Kamps, W.A.; et al. Prognostic value of minimal residual disease in acute lymphoblastic leukaemia in childhood. Lancet 1998, 352, 1731–1738. [Google Scholar] [CrossRef]
  83. Conter, V.; Bartram, C.R.; Valsecchi, M.G.; Schrauder, A.; Panzer-Grümayer, R.; Möricke, A.; Aricò, M.; Zimmermann, M.; Mann, G.; De Rossi, G.; et al. Molecular response to treatment redefines all prognostic factors in children and adolescents with B-cell precursor acute lymphoblastic leukemia: Results in 3184 patients of the AIEOP-BFMALL 2000 study. Blood 2010, 115, 3206–3214. [Google Scholar] [CrossRef]
  84. Strati, P.; Keating, M.J.; O’Brien, S.M.; Burger, J.; Ferrajoli, A.; Jain, N.; Tambaro, F.P.; Estrov, Z.; Jorgensen, J.; Challagundla, P.; et al. Eradication of bone marrow minimal residual disease may prompt early treatment discontinuation in CLL. Blood 2014, 123, 3727–3732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Santacruz, R.; Villamor, N.; Aymerich, M.; Martínez-Trillos, A.; López, C.; Navarro, A.; Rozman, M.; Beà, S.; Royo, C.; Cazorla, M.; et al. The prognostic impact of minimal residual disease in patients with chronic lymphocytic leukemia requiring first-line therapy. Haematologica 2014, 99, 873–880. [Google Scholar] [CrossRef] [Green Version]
  86. Logan, A.C.; Zhang, B.; Narasimhan, B.; Carlton, V.; Zheng, J.; Moorhead, M.; Krampf, M.R.; Jones, C.D.; Waqar, A.N.; Faham, M.; et al. Minimal residual disease quantification using consensus primers and high-throughput IGH sequencing predicts post-transplant relapse in chronic lymphocytic leukemia. Leukemia 2013, 27, 1659–1665. [Google Scholar] [CrossRef] [PubMed]
  87. Kwok, M.; Rawstron, A.C.; Varghese, A.; Evans, P.A.S.; O’Connor, S.J.M.; Doughty, C.; Newton, D.J.; Moreton, P.; Hillmen, P. Minimal residual disease is an independent predictor for 10-year survival in CLL. Blood 2016, 128, 2770–2773. [Google Scholar] [CrossRef] [Green Version]
  88. Kovacs, G.; Robrecht, S.; Fink, A.M.; Bahlo, J.; Cramer, P.; Von Tresckow, J.; Maurer, C.; Langerbeins, P.; Fingerle-Rowson, G.; Ritgen, M.; et al. Minimal residual disease assessment improves prediction of outcome in patients with chronic lymphocytic Leukemia (CLL) who achieve partial response: Comprehensive analysis of two phase III Studies of the German CLL Study Group. J. Clin. Oncol. 2016, 34, 3758–3765. [Google Scholar] [CrossRef]
  89. Dimier, N.; Delmar, P.; Ward, C.; Morariu-Zamfir, R.; Fingerle-Rowson, G.; Bahlo, J.; Fischer, K.; Eichhorst, B.; Goede, V.; Van Dongen, J.J.M.; et al. A model for predicting effect of treatment on progression-free survival using MRD as a surrogate end point in CLL. Blood 2018, 131, 955–962. [Google Scholar] [CrossRef] [PubMed]
  90. Böttcher, S.; Ritgen, M.; Fischer, K.; Stilgenbauer, S.; Busch, R.M.; Fingerle-Rowson, G.; Fink, A.M.; Buḧler, A.; Zenz, T.; Wenger, M.K.; et al. Minimal residual disease quantification is an independent predictor of progression-free and overall survival in chronic lymphocytic leukemia: A Multivariate analysis from the randomized GCLLSG CLL8 trial. J. Clin. Oncol. 2012, 30, 980–988. [Google Scholar] [CrossRef]
  91. Paiva, B.; Puig, N.; Cedena, M.T.; Rosiñol, L.; Cordón, L.; Vidriales, M.B.; Burgos, L.; Flores-Montero, J.; Sanoja-Flores, L.; Lopez-Anglada, L.; et al. Measurable residual disease by next-generation flow cytometry in multiple myeloma. J. Clin. Oncol. 2020, 38, 784–792. [Google Scholar] [CrossRef]
  92. Lahuerta, J.J.; Paiva, B.; Vidriales, M.B.; Cordón, L.; Cedena, M.T.; Puig, N.; Martinez-Lopez, J.; Rosiñol, L.; Gutierrez, N.C.; Martín-Ramos, M.L.; et al. Depth of response in multiple myeloma: A pooled analysis of three PETHEMA/GEM clinical trials. J. Clin. Oncol. 2017, 35, 2900–2910. [Google Scholar] [CrossRef] [Green Version]
  93. Rawstron, A.C.; Child, J.A.; De Tute, R.M.; Davies, F.E.; Gregory, W.M.; Bell, S.E.; Szubert, A.J.; Navarro-Coy, N.; Drayson, M.T.; Feyler, S.; et al. Minimal residual disease assessed by multiparameter flow cytometry in multiple myeloma: Impact on outcome in the Medical Research Council Myeloma IX study. J. Clin. Oncol. 2013, 31, 2540–2547. [Google Scholar] [CrossRef] [PubMed]
  94. Munshi, N.C.; Avet-Loiseau, H.; Rawstron, A.C.; Owen, R.G.; Child, J.A.; Thakurta, A.; Sherrington, P.; Samur, M.K.; Georgieva, A.; Anderson, K.C.; et al. Association of Minimal Residual Disease With Superior Survival Outcomes in Patients With Multiple Myeloma: A Meta-analysis. JAMA Oncol. 2017, 3, 28. [Google Scholar] [CrossRef] [PubMed]
  95. Szczepański, T. Why and how to quantify minimal residual disease in acute lymphoblastic leukemia? Leukemia 2007, 21, 622–626. [Google Scholar] [CrossRef] [Green Version]
  96. Rawstron, A.C.; Kennedy, B.; Evans, P.A.S.; Davies, F.E.; Richards, S.J.; Haynes, A.P.; Russell, N.H.; Hale, G.; Morgan, G.J.; Jack, A.S.; et al. Quantitation of minimal disease levels in chronic lymphocytic leukemia using a sensitive flow cytometric assay improves the prediction of outcome and can be used to optimize therapy. Blood 2001, 98, 29–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Theunissen, P.; Mejstrikova, E.; Sedek, L.; Van Der Sluijs-Gelling, A.J.; Gaipa, G.; Bartels, M.; Sobral da Costa, E.; Kotrová, M.; Novakova, M.; Sonneveld, E.; et al. Standardized flow cytometry for highly sensitive MRD measurements in B-cell acute lymphoblastic leukemia. Blood 2017, 129, 347–357. [Google Scholar] [CrossRef]
  98. Van Dongen, J.J.M.; Langerak, A.W.; Brüggemann, M.; Evans, P.A.S.; Hummel, M.; Lavender, F.L.; Delabesse, E.; Davi, F.; Schuuring, E.; García-Sanz, R.; et al. Design and standardization of PCR primers and protocols for detection of clonal immunoglobulin and T-cell receptor gene recombinations in suspect lymphoproliferations: Report of the BIOMED-2 concerted action BMH4-CT98-3936. Leukemia 2003, 17, 2257–2317. [Google Scholar] [CrossRef] [Green Version]
  99. Brüggemann, M.; Kotrová, M.; Knecht, H.; Bartram, J.; Boudjogrha, M.; Bystry, V.; Fazio, G.; Froňková, E.; Giraud, M.; Grioni, A.; et al. Standardized next-generation sequencing of immunoglobulin and T-cell receptor gene recombinations for MRD marker identification in acute lymphoblastic leukaemia; a EuroClonality-NGS validation study. Leukemia 2019, 33, 2241–2253. [Google Scholar] [CrossRef] [Green Version]
  100. Van der Velden, V.H.J.; Cazzaniga, G.; Schrauder, A.; Hancock, J.; Bader, P.; Panzer-Grumayer, E.R.; Flohr, T.; Sutton, R.; Cave, H.; Madsen, H.O.; et al. Analysis of minimal residual disease by Ig/TCR gene rearrangements: Guidelines for interpretation of real-time quantitative PCR data. Leukemia 2007, 21, 604–611. [Google Scholar] [CrossRef] [Green Version]
  101. Kotrova, M.; Van Der Velden, V.H.J.; Van Dongen, J.J.M.; Formankova, R.; Sedlacek, P.; Brüggemann, M.; Zuna, J.; Stary, J.; Trka, J.; Fronkova, E. Next-generation sequencing indicates false-positive MRD results and better predicts prognosis after SCT in patients with childhood ALL. Bone Marrow Transplant. 2017, 52, 962–968. [Google Scholar] [CrossRef] [Green Version]
  102. Cavalli, M.; De Novi, L.A.; Della Starza, I.; Cappelli, L.V.; Nunes, V.; Pulsoni, A.; Del Giudice, I.; Guarini, A.; Foà, R. Comparative analysis between RQ-PCR and digital droplet PCR of BCL2/IGH gene rearrangement in the peripheral blood and bone marrow of early stage follicular lymphoma. Br. J. Haematol. 2017, 177, 588–596. [Google Scholar] [CrossRef] [Green Version]
  103. Drandi, D.; Kubiczkova-Besse, L.; Ferrero, S.; Dani, N.; Passera, R.; Mantoan, B.; Gambella, M.; Monitillo, L.; Saraci, E.; Ghione, P.; et al. Minimal Residual Disease Detection by Droplet Digital PCR in Multiple Myeloma, Mantle Cell Lymphoma, and Follicular Lymphoma: A Comparison with Real-Time PCR. J. Mol. Diagn. 2015, 17, 652–660. [Google Scholar] [CrossRef]
  104. Della Starza, I.; Nunes, V.; Cavalli, M.; De Novi, L.A.; Ilari, C.; Apicella, V.; Vitale, A.; Testi, A.M.; Del Giudice, I.; Chiaretti, S.; et al. Comparative analysis between RQ-PCR and digital-droplet-PCR of immunoglobulin/T-cell receptor gene rearrangements to monitor minimal residual disease in acute lymphoblastic leukaemia. Br. J. Haematol. 2016, 174, 541–549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Della Starza, I.; Nunes, V.; Lovisa, F.; Silvestri, D.; Cavalli, M.; Garofalo, A.; Campeggio, M.; De Novi, L.A.; Soscia, R.; Oggioni, C.; et al. Droplet Digital PCR Improves IG-/TR-based MRD Risk Definition in Childhood B-cell Precursor Acute Lymphoblastic Leukemia. HemaSphere 2021, 5, e543. [Google Scholar] [CrossRef]
  106. Hansen, M.H.; Cédile, O.; Larsen, T.S.; Abildgaard, N.; Nyvold, C.G. Perspective: Sensitive detection of residual lymphoproliferative disease by clonal gene rearrangements and NGS–How low can you go? Exp. Hematol. 2021, 98, 14–24. [Google Scholar] [CrossRef] [PubMed]
  107. Knecht, H.; Reigl, T.; Kotrová, M.; Appelt, F.; Stewart, P.; Bystry, V.; Krejci, A.; Grioni, A.; Pal, K.; Stranska, K.; et al. Quality control and quantification in IG/TR next-generation sequencing marker identification: Protocols and bioinformatic functionalities by EuroClonality-NGS. Leukemia 2019, 33, 2254–2265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Low- and high-throughput (NGS) IG/TR analyses in different sample types. (A) In samples from healthy individuals, NGS provides sequences of all clones. These can also be obtained by low-throughput Sanger sequencing, but it is necessary to clone the PCR products before sequencing to obtain a clean sequence of each clone separately. (B) In highly infiltrated diagnostic samples, NGS provides a sequence of the dominant malignant clone (red) and background clones, as well as information on their abundance. Sanger sequencing provides a sequence of the dominant rearrangement only, without abundance information. (C) In samples after treatment, NGS can detect the rearrangement of the malignant clone (red) and the background rearrangements. RQ-PCR or ddPCR only provide the information about the MRD level.
Figure 1. Low- and high-throughput (NGS) IG/TR analyses in different sample types. (A) In samples from healthy individuals, NGS provides sequences of all clones. These can also be obtained by low-throughput Sanger sequencing, but it is necessary to clone the PCR products before sequencing to obtain a clean sequence of each clone separately. (B) In highly infiltrated diagnostic samples, NGS provides a sequence of the dominant malignant clone (red) and background clones, as well as information on their abundance. Sanger sequencing provides a sequence of the dominant rearrangement only, without abundance information. (C) In samples after treatment, NGS can detect the rearrangement of the malignant clone (red) and the background rearrangements. RQ-PCR or ddPCR only provide the information about the MRD level.
Genes 12 00979 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kotrova, M.; Darzentas, N.; Pott, C.; Baldus, C.D.; Brüggemann, M. Immune Gene Rearrangements: Unique Signatures for Tracing Physiological Lymphocytes and Leukemic Cells. Genes 2021, 12, 979. https://doi.org/10.3390/genes12070979

AMA Style

Kotrova M, Darzentas N, Pott C, Baldus CD, Brüggemann M. Immune Gene Rearrangements: Unique Signatures for Tracing Physiological Lymphocytes and Leukemic Cells. Genes. 2021; 12(7):979. https://doi.org/10.3390/genes12070979

Chicago/Turabian Style

Kotrova, Michaela, Nikos Darzentas, Christiane Pott, Claudia D. Baldus, and Monika Brüggemann. 2021. "Immune Gene Rearrangements: Unique Signatures for Tracing Physiological Lymphocytes and Leukemic Cells" Genes 12, no. 7: 979. https://doi.org/10.3390/genes12070979

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