Open access peer-reviewed chapter

Clonality Testing in the Diagnosis of Cutaneous T-Cell Lymphoma

Written By

Fernando Roncolato, Anna Gazzola, Claudia Mannu, Riccardo Donelli, Maryam Etebari, Mohsen Navari and Pier Paolo Piccaluga

Submitted: 18 January 2024 Reviewed: 18 January 2024 Published: 10 April 2024

DOI: 10.5772/intechopen.1004283

From the Edited Volume

Molecular Diagnostics of Cancer

Pier Paolo Piccaluga

Chapter metrics overview

6 Chapter Downloads

View Full Metrics

Abstract

Cutaneous T-cell lymphomas (CTCL) are heterogeneous disorders including several different entities with variable morphology, phenotype, and clinical features. The diagnosis of CTCL is often challenging; the distinction between tumors and reactive, non-neoplastic conditions is sometimes elusive. Therefore, molecular testing for T-cell receptor gamma (TRG) clonality is often performed. In this study, we evaluated the accuracy of TRG testing protocol in 110 routinary cases and discussed the subject of clonality testing in light of novel technologies (namely next-generations sequencing advent). For TRG analysis, the BIOMED-2 protocol was adopted. Sensitivity, specificity, positive and negative predictive values were calculated using the Oxford CatMaker software following the Standards for Reporting of Diagnostic Accuracy Studies (STARD) requirements for diagnostic accuracy. We found that this approach was feasible in most cases (87%) despite the small sample dimensions and the fixation issues. In addition, we found that sensitivity and specificity were 90 and 84%, respectively; accordingly, positive predictive value (PPV) and negative predictive value (NPV) were 84 and 90%, respectively. Of note, the molecular test was somehow influential in 83% of cases, when histology and phenotyping were not conclusive. In conclusion, TRG analysis by BIOMED-2 protocols is feasible and effective in the routine diagnostics of CTCL. The integration of histological, phenotypical, molecular, and clinical data is mandatory for a correct diagnosis.

Keywords

  • T-cell receptor gamma
  • T-cell lymphoma
  • cutaneous T-cell lymphoma
  • BIOMED-2
  • clonality
  • diagnostic accuracy
  • evidence-based medicine
  • next-generation sequencing
  • molecular diagnostic
  • PCR

1. Introduction

Peripheral T-cell lymphomas (PTCLs) are clonal neoplasms deriving from mature T-lymphocytes at different stages of differentiation [1]. Clinical presentations are protean, and diagnosis involves a synthesis of clinical history and pathological data from morphological, immunophenotypic, cytogenetic, and molecular investigations. Based on such criteria, the recent WHO Classification of Tumors of Hematopoietic and Lymphoid Tissues codified many disease entities, identifying specified and not otherwise specified (NOS) forms, nodal and extranodal tumors [1]. Among the latter, cutaneous T-cell lymphomas (CTCL) are relatively common, including several distinct diseases [2]. The diagnosis of CTCL, as for other PTCLs, is challenging owing to the heterogeneous pathophysiology, complex taxonomy, and, especially, the lack of tumor-associated immunophenotypic or molecular markers. Furthermore, the clinical scenario becomes yet more complicated when the disease presents in the early phase or with the contemporaneous occurrence of a prominent reactive component overwhelming the malignant elements [2].

Nonetheless, importantly, the diagnosis of lymphoproliferative disease can benefit from the demonstration of clonality at a molecular level. Specifically, in the field of T-cell lymphomas/leukemias, the study of T-cell receptor gene (TCR) rearrangements has been used to assist in the determination of clonality for over two decades [3, 4]. The somatic assembly of TCR genes is an early event during T-cell development, occurring through a V(D)J recombination directed by site-specific recombinase (RAG1 and RAG2), which eventually leads to a diverse T-cell population [3, 5]. In 2003, the BIOMED-2 consortium established standardized protocols for multiplex PCR analysis, which improved the efficiency and reproducibility of clonality detection [6]. This protocol was validated by the BIOMED-2 group in 2007 in 188 cases of T-cell malignancy [7, 8]. Specifically, the combined analysis of TCR-gamma (TRG) and TCR-beta (TRB) led to sensitivity and specificity values above 90% [6, 7, 8].

Although the efficiency of BIOMED-2 protocols was defined, in both studies, the BIOMED-2 protocol was tested in a series of tissues with well-established histological diagnoses of T-cell malignancy, principally represented by fresh or frozen specimens [6, 7, 8]. By contrast, routine clinical diagnosis is predominantly based on formalin-fixed paraffin-embedded (FFPE) tissue blocks. FFPE blocks are convenient as they are small, easy to transport, and allow for retrospective analysis by immunohistochemistry and in situ hybridization (ISH/FISH). However, by comparison to fresh specimens, molecular analyses on FFPE blocks are more challenging owing principally to the more involved method required for extracting high-quality DNA. The interference of formalin fixation with DNA architecture and nucleoside fragmentation can lead to lower amplification yield and potentially a lower diagnostic utility [9, 10]. In this regard, small specimens, particularly those such as skin biopsies, represent a significant challenge in molecular testing. Practically, the two steps (TRG and TRB) can be performed simultaneously to save time, or consecutively, to save resources. In fact, the latter solution may allow the analysis to be limited to TRG in most instances. Noteworthy is that a formal, evidence-based diagnostic accuracy study has never been performed in this setting.

In order to properly quantify this possibility, in this study we performed a diagnostic accuracy study based on a retrospective analysis of our experience with the BIOMED-2 TRG protocol in 110 skin specimens aiming to: [1] assess the feasibility of the protocol in non-selected consecutive FFPE cases included in the routine practice; [2] assess sensitivity, specificity, PPV and NPV of the test in detecting T-cell malignancies in FFPE tissues; and [3] determine the rate and clinical significance of clonal rearrangements in cases deemed to be reactive or non-diagnosable by histopathological and morphological assessment (“TRG monoclonality of uncertain significance”).

Furthermore, we discuss the issue of clonality testing in light of the new available technologies, namely next-generation sequencing (NGS).

Advertisement

2. Materials and methods

2.1 Case selection

We included in this study 110 consecutive cases received over 3 years at the Molecular Pathology Laboratory, Hematopathology Unit, Institute of Hematology and Medical Oncology “L. & A. Seràgnoli.” Histological samples were classified by at least two experienced hematopathologists who were blinded to the TRG result using the morphological, immunohistochemical, and cytogenetic criteria of the most updated WHO classification [2]. The diagnoses were as follows: PTCL/CTCL (N = 48; 49%), B-cell lymphomas (N = 8; 8%), and inflammatory disorders (N = 42, 43%). We also studied a further 12 cases for which a definitive diagnosis was not reached based on morphology, phenotype, or clinical presentation, and where the differential between tumor and inflammatory disorders was not resolved. For all cases, a minimum of 12 months of clinical follow-up from the last PCR study was available. Patients’ characteristics are reported in Table A1.

All specimens were formalin-fixed and paraffin-embedded (FFPE) prior to PCR analysis.

Calculations of sensitivity (ST), specificity (SP), positive predictive value (PPV), and negative predictive value (NPV) were made by CATmaker software (Centre for Evidence-Based Medicine, Oxford University, http://www.cebm.net) using the blinded histopathological review as the reference test in our case series.

For the purposes of this analysis, a distinction was not drawn between primary cutaneous T-cell lymphomas and systemic lymphomas manifesting in the skin, as the analysis was designed to assess the capacity of the molecular investigation to detect any T-cell lymphoma in the skin. The STARD Statement for diagnostic accuracy studies was fulfilled (Table A2) [11].

The study design is summarized in Figure 1.

Figure 1.

Schematic representation of the diagnostic accuracy study design.

2.2 Molecular analysis

Molecular tests were performed and analyzed by at least two experienced molecular pathologists. Molecular testing was performed within 1 week of the histological evaluation. Histopathological evaluation and molecular tests were performed in a blinded manner.

Genomic DNA was extracted from paraffin-embedded tissue by using a Qiagen DNA mini kit according to the manufacturer’s instructions. PLZF was studied as a control gene in order to verify the DNA integrity. PCR analysis of TRG rearrangement was executed using multiplex PCR tubes (BIOMED-2 Concerted Action) [6]. Amplification reactions were performed in an automated thermocycler (mastercycler Eppendorf) according to the BIOMED-2 multiplex PCR protocol [6]. Each 50-microliter PCR reaction included 100 ng of DNA, 10 pm of each primer, 0,2 mmol/L dNTP, 5 microliter of 10X Gold buffer, 1.5 mmol/L of MgCl2 and 1 U of Ampli-Taq Gold polymerase (Applied Biosystems). The cycling parameters were as follows: pre-activation for 7 minutes at 95°C, 35 cycles of denaturation (95°C for 30 sec), an annealing step (60°C for 30 sec), an extension step (72°C for 60 sec), finally 10 minutes of extension at 72°C. Each sample was evaluated using heteroduplex analysis and GeneScanning to determine polyclonal or monoclonal character.

2.2.1 Heteroduplex analysis

In order to allow the spontaneous formation of homoduplex or heteroduplex from DNA fragments, amplicons were denatured by heating (94°C for 5 min) and cooled at low temperatures (4°C for 1 hour). PCR products were then visualized by electrophoresis on polyacrylamide gels being separated according to their length and conformation. According to BIOMED-2 protocols, samples were defined as monoclonal when a single band was identified within a predictable size range and polyclonal when only a smear was detected [6]. This technique has a well-recognized detection limit of ∼5% due to the frequency of polyclonal/reactive lymphocytes present in the tissue.

2.2.2 GeneScanning analysis

After amplification, 1 μ of PCR product with 0.5 μ of a standard molecular weight product (LIZ Applied Biosystems) was mixed with 12 μ of formamide to induce denaturation into single DNA strands (1 minute to 95C°). Subsequently, they were separated through a polymer capillary electrophoresis system and automatically detected by fluorescence reading with a laser system in an automatic DNA sequencer (ABI Prism 310 Applied Biosystems).

Tissue samples were considered to have a clonal T-cell population if 1 or 2 peaks of the amplified PCR product in question were obtained (monoclonal and biclonal/biallelic, respectively); detection of three to five peaks was counted as an oligoclonal result, while a Gaussian distribution of peaks was referred to as a polyclonal population [6]. The control gene minimum amplification requirement was 300 bps, and when this was not reached, or the target gene was not amplified, the sample was considered “not evaluable.”

Advertisement

3. Results

A total of 110 case samples were interrogated using the BIOMED-2 protocol for TRG analysis. Fourteen cases did not have an evaluable molecular result (14/110, 12.7%), owing to failure to amplify either the target gene product or the control gene, and were then excluded from the final analysis. 96/110 samples (87.3%) could be evaluated and were included in the final analysis.

Of the specimens that allowed a clear and definitive histopathological diagnosis, 84/96 had an evaluable molecular result (87.5%).

Based on TRG analysis, there was evidence of clonal rearrangement in 42/84 (50%) of cases, oligoclonal in 3/84 (4%), and a polyclonal pattern was detected in 39/84 (46%) cases, respectively.

Mono/biclonality was detected in 36/47 (77%) of the CTCL cases, 2/8 (25%) of the B-NHL cases, and 4/42 (10%) of the inflammatory reactions.

The ST and SP of the molecular test in discriminating malignant T-cell lymphoproliferation vs. other conditions (including B-NHL and inflammatory reactions) were 90.0 and 84%, respectively. Accordingly, PPV and NPV were 84.0 and 90%, respectively (Table 1).

Target: Differential diagnosis CTCL vs. Other
Histopathological diagnosis
CTCLOther
TRGClonal vs. Oligo-polyclonal367
437
ParameterValue95% CI
ST90%81–99
SP84%73–95
Pre-test probability48%37–58
PPV84%73–95
NPV90%81–99
LR+5.662.85 to 11.25
LR-0.120.05–0.30
ST = sensitivity
SP = specificity
PPV = positive predictive value
NPV = negative predictive value
LR + = likelyhood ratio positive
LR- = likelyhood ratio negative

Table 1.

Calculation of sensitivity (ST), specificity (SP), positive predictive value (PPV) and negative predictive value (NPV), basing on the histopathological analysis taken as diagnostic gold standard.

CATmaker software (Centre for Evidence-Based Medicine, Oxford University, http://www.cebm.net).

Subsequently, we evaluated the 12 cases for which a conclusive diagnosis was not made by histopathological review. Specifically, we detected monoclonal rearrangement in 4/12 cases, oligoclonal rearrangement in 3/12, and 5/12 presented with a polyclonal pattern. In the monoclonal group, 3/4 ultimately received a diagnosis of CTCL after the integration of all diagnostic procedures. Among oligoclonal/polyclonal cases, 1 was eventually diagnosed as CTCL, while the remaining 8 were diagnosed as inflammatory conditions. Overall, based on such data, the molecular result was influential on the final diagnosis in 10/12 (83%) of the cases when morphology, immunophenotype, and molecular data were integrated (Table 2).

Number of casesDiagnosis
TCLNon-TCL
TRG
Monoclonal43 (75%)1 (25%)
polyclonal/oligoclonal81/0 (12.5%)4/3 (87.5%)

Table 2.

Impact of the molecular result in cases with undefined diagnosis.

We then evaluated the molecular results in PTCLs according to the histological subtype in order to assess whether specific diseases are differentially associated with different degrees of clonality at TRG analysis (Table 3). We found that there were in fact no significant differences recorded, with TRG successful in detecting clonal rearrangements in the majority of cases (Table 4).

Lymphoma typeNumber of casesEvidence of clonality (number of cases)Evidence of clonality (percentage of cases)
Mycosis fungoides181688.89
PTCL/NOS151386.67
CTCL/NOS55100.00
T-LGL11100.00
ALCL, ALK-11100.00
Total403690.00

Table 3.

TRG clonality according to lymphoma histotype.

SampleTissueEfficiency (% of evaluable cases)ProbeST (%)SP (%)PPV (%)NPV (%)
SeràgnoliFFPESkin88γ73807578
ZhangFFPESkinn/aγ6484varvar
β6484varvar
γ + β7874varvar
LukowskyFFPESkinn/aγ81n/an/an/a
β78n/an/an/a
γ + β87n/an/an/a
GoeldelFrozenSkin0γ77848479
PontiFresh/frozenSkinn/aγ84989592
Biomed-2 GroupFresh/frozenNot specifiedn/aγ89n/an/an/a
β91n/an/an/a
γ + β94n/an/an/a

Table 4.

Comparison of sensitivity (ST), specificity (SP), positive predictive value (PPV), and negative predictive value (NPV) in different studies adopting the BIOMED-2 protocols.

n/a = not available.

var = variable PPV and NPV dependent on pre-test probability.

Finally, we investigated whether cases presenting evidence of clonal rearrangement but without clear evidence of PTCL had a higher risk of developing T-cell malignancies in the following months. Specifically, among cases that received a final diagnosis of inflammatory disorder, 4/42 (9.5%) demonstrated TRG clonality. These particular patients largely fulfilled the minimum requested follow-up of 12 months, the mean follow-up period being 35 months (range 33–36 months). Patients were then monitored by means of outpatient clinical follow-up and/or phone contact with the treating physician. No patient received a diagnosis of a T-cell neoplasm during the study period, to the best of our knowledge.

Matched histological and molecular analysis results are detailed in Table A3.

Advertisement

4. Discussion

In this study, we examined the sensitivity and specificity and, accordingly, PPV and NPV, of the BIOMED-2 protocol in detecting TRG clonal rearrangements in cutaneous T-cell lymphoproliferative disorders. We included 110 consecutive cases of histologically complex FFPE skin biopsies referred to our institution over a 3-year period.

In our hands, the BIOMED-2 TRG protocol was technically successful in 87% of cases, based on a control gene minimum amplification requirement of 300 bps. In particular, it demonstrated a sensitivity of 90.0% and a specificity of 84%. The sensitivity and specificity data using this same approach have been published in previous papers (Table 4), and the figures obtained in our institution are comparable to previously published data in FFPE specimens. This suggests that sensitivity in FFPE specimens can approach that of fresh frozen specimens. With regard to FFPE samples, Zhang et al. reported a sensitivity and specificity of 64 and 84%, respectively, in a case series looking exclusively at MF, while Lukowsky et al. reported a sensitivity of 81% [12, 13]. In fresh/frozen tissues, Goeldel et al. reported a sensitivity and specificity of 77 and 84%, respectively [9, 14], while Ponti et al. described slightly higher figures of 83.5 and 97.7%, respectively [15]. A direct comparison with the original data from the BIOMED-2 consortium does, however, pose some problems as that group only analyzed fresh and frozen specimens and included non-skin specimens as well (Table 4).

Our study differs from the previous one, being the first phase 3 diagnostic accuracy study specifically designed and conducted according to the STRAD requirements for evidence-based medicine [11]. In doing so, we evaluated, for the first time in the specific setting of FFPE samples, the PPV and NPV of the BIOMED-2 TRG protocol. Importantly, such variables are strictly dependent on the incidence of the disease in the population tested (pre-test probability). In our laboratory, the use of TRG rearrangement testing is typically reserved for those cases where diagnostic uncertainty persists after morphological assessment with immunohistochemical staining, in which the pre-test probability is moderate/high and in which a positive or negative result may influence the final diagnosis [16]. In this context, the addition of a molecular tool as a diagnostic aid becomes significant. In this series, the pre-test probability was quite balanced, being 48%. We calculated a PPV of 84% with an NPV of 90% and found that most histologically “uncertain” cases could be satisfactorily resolved with the addition of the PCR information. Recently, Zhang et al. created a model that demonstrated the effect of pre-test probability on the PPV and NPV and suggested that the molecular result should only be considered when the pre-test probability lies between 0.15 and 0.75 [12], and this is actually consistent with our practice. Zhang et al. further pointed out that at the extremes of the pre-test probability range, the molecular result should be ignored [12]. Practically, based on the same SP and ST recorded in our study, a pre-test probability of 8% would lead to a positive predictive value of as low as 34%. Indeed, this underlines the importance of patient selection based on morphology and phenotype before molecular testing, which cannot represent a reliable screening test.

Regarding fresh/frozen samples, Ponti et al. described a remarkable PPV and NPV of 95 and 92%, respectively. It should be noted, however, that PPV and PNV may vary in the different studies due to differences in the considered series (i.e., research vs. routine diagnostic). In fact, at times, the clinical imperative urges contemporaneous initiation of multiple investigative strategies, including PCR, not respecting the ideal logical order (histology, immunohistochemistry, and genetics), thus affecting the pre-test probability. This real-world environment may alter the PPV and the NPV of the analysis. Further, the addition of TRB and TRD analysis, not available in this series, would potentially certainly improve the efficiency of the BIOMED-2 protocol. In this regard, it should be noted that this study did not aim to assess the accuracy of the entire protocol but rather of the TRG step, which is adopted as the first and often only one in many laboratories. For sure, the addition of the subsequent steps would increase the overall sensitivity, though negatively affecting, on the other hand, the specificity.

As mentioned, in spite of these remarkable figures for PPV and NPV, the importance of clinicopathologic integration in making a diagnosis in such a complex clinical setting must be remembered. It should be kept in mind that clonality, per sè, does not always indicate malignancy, and it is well-recognized that clonality can also be seen in reactive processes and thus cannot be considered a “sine qua non” of cancer [14, 17, 18, 19, 20]. However, when clonality is detected without a definitive diagnosis of malignancy, a close clinical follow-up is mandatory, in order to ensure occult T-cell lymphoma is not missed [17, 21, 22]. In fact, progression to frank T-cell malignancy has been reported with variable frequency [23, 24]. Indeed, several studies have examined the fate of patients with an indeterminate diagnosis, with a variable percentage of patients who later on presented with a clear CTCL, ranging from 0 to 85% [15, 25, 26, 27]. With this in mind, we followed up on cases that demonstrated clonality, but the global picture was not indicative of lymphoma. However, in our experience, no one developed a lymphoma within the follow-up period. We cannot exclude that a later onset might occur.

Diagnostic algorithms for MF incorporating molecular testing with integrated clinicopathologic findings have been examined by a number of groups including most notably the ISCL [28, 29, 30, 31]. Our findings suggested that TRG performed in FFPE in this subgroup remains a valid method of supporting a diagnosis in histologically challenging cases. Particularly, we had evidence that TRG clonal rearrangements were clinically useful also when a clear diagnosis of lymphoma could not be established. The role of molecular clonality testing in influencing the diagnosis of cutaneous lymphoid pathology in cases that are histologically uncertain needs further investigation. Nevertheless, it is now considered part of the diagnostic repertoire available to the pathologist and, indeed, has formed part of the ISCL and EORTC guidelines for diagnosis of MF since 2007 [29]. In our study, the most common malignancy identified was MF, in which 16/18 cases (88.9%) demonstrated TRG clonality. This compares to 68.2–88% as reported in the literature [12, 30], possibly reflecting the remarkable selection of cases based on immunomorphology in dedicated and ultra-specialistic Hematopathology Units. It is conceivable that the addiction of miRNA analysis [32] as well as comprehensive genetic testing [33, 34, 35], will further increase the importance of molecular pathology in CTCL analysis.

Finally, recent studies showed the potential efficacy of a next-generation sequencing (NGS) approach to resolve clonality testing [36]. These methods are based on the preparation of a DNA samples library amplified by PCR, using appropriate forward and reverse primers (tagged with unique barcodes) which usually target, respectively, different portions of V and J regions of the TRG gene; the amplified library is subsequently sequenced through a reversible dye-terminators technique and analyzed with bioinformatics approaches that return the proportion of the given sequences and assign them a rearrangement identity based on an alignment score [37, 38, 39]. These technologies are complex and parallelized processes that permit the simultaneous analysis of several samples returning high-throughput data. The introduction of NGS approach for the clonality assessment in CTCL can lead to useful advantages with respect to the gold standard PCR-based ones, overcoming its main limits [40, 41]: first, the possibility of distinguishing between same-sized amplified sequences, given that sequencing separates them also based on their nucleotides composition and not only on their length; further, also the possibility to better interpreting ambiguous results, as non-uniform Gaussian distributions, and better resolving a polyclonal background. In addition, this approach necessitates a minor amount of DNA (10–20 ng vs. 100–500 ng) and has the ability to detect and quantify also recurrent minor clones with small numbers of circulating tumor cells: as a result, NGS can improve the monitoring of the disease progression and treatments response, especially regarding minimal residual disease (MRD) context [42]. Hence, these new techniques could lead to a better and more objective classification, stratification, and monitoring of lymphoid malignancies, such as CTCL, which are essential proprieties in molecular diagnostics.

Advertisement

5. Conclusions

In conclusion, the TRG BIOMED-2 protocol appeared to be a feasible and remarkably effective method for analyzing clonality from FFPE specimens in histologically challenging cases. However, as a slight overestimation of clonal restrictions is possible, analysis repetition and careful patient follow-up are prudent. Proper clinical comparison with NGS technologies [36] is now certainly warranted.

Advertisement

Acknowledgments

The authors are grateful to Dr. Francesco Bacci, Dr. Elena Sabattini, and Dr. Carlo Sagramoso-Sacchetti for their contribution to diagnostic assessments as well as to Prof. Stefano A. Pileri for the precious advice.

The work reported in this publication was funded by the Italian Ministry of Health, RC-2023-2778976 (Prof. Piccaluga) and FIRB Futura 2011 RBFR12D1CB.

Advertisement

Conflict of interest

“The authors declare no conflict of interest.”

Advertisement

Note

Prof. Pier Paolo Piccaluga is currently affiliated with the Jomo Kenyatta University of Agriculture and Technology (Nairobi, Kenya), The University of Nairobi (Nairobi, Kenya), and the University of Botswana (Gaborone, Botswana).

Advertisement

Appendix

Case numberGenderSite of biopsyClinical History
1MskinSkin lesion NOS
2MskinSkin lesion NOS
3FskinSkin lesion NOS
4FskinSkin lesion in pregressed NHL
5MskinSkin lesion NOS
6MskinSkin lesion NOS
7MskinErythema
8FskinDermatosis
9MskinPsoriasis + adenopathies
10MskinSkin lesion NOS
11FskinTumoral skin lesion
12FskinHashimoto thyroiditis
13MskinAbdominal erythema
14MskinSkin lesion NOS
15MskinSkin nodules at lower limbs
16FskinSkin lesion NOS
17MskinSkin lesion NOS
18MskinSkin lesion NOS
19MskinSuspected Lymphomatoid Papulosis
20MskinSkin lesion NOS
21FskinScalp erythema
22MskinSkin lesion in pregressed NHL
23FskinSkin lesion NOS
24FskinSkin lesion NOS
25FskinSkin lesion NOS
26FskinSkin lesion NOS
27MskinSkin lesion NOS
28MskinSkin lesion NOS
29FskinErythema
30MskinErythema
31MskinPapulo-nodular skin lesions
32MskinSuspected lymphoma
33FskinCutaneous pseudolymphoma
34FskinPlaque
35FskinSkin lesion NOS
36MskinMycosis fungoides
37FskinErythrodermic lesion
38MskinSkin lesion NOS
39MskinSkin lesion NOS
40FskinSkin lesion NOS
41MskinSkin lesion NOS
42FskinSkin lesion NOS
43MskinSkin lesion NOS
44FskinSkin lesion NOS
45MskinErythema
46MskinLower limb ulcer
47FskinSkin lesion NOS
48FskinSuspected insect bite
49MskinSkin lesion NOS
50FskinSkin lesion NOS
51MskinSkin lesion NOS
52MskinPlaque
53MskinSkin lesion + adenopathy
54MskinPlaque
55MskinSkin lesion NOS
56FskinSkin lesion NOS
57FskinSkin lesion NOS
58FskinTumoral skin lesion
59FskinSkin lesion NOS
60FskinMycosis fungoides
61MskinSkin lesions
62MskinTumoral skin lesion
63MskinMycosis fungoides
64MskinSkin lesion NOS
65MskinSkin lesion NOS
66FskinHIV+ + skin lesions
67MskinSkin lesion NOS
68FskinSkin lesion NOS
69MskinSkin lesion NOS
70FskinTumoral skin lesion
71MskinSkin lesion NOS
72MskinSkin lesion NOS
73MskinSkin lesion NOS
74MskinSkin lesion NOS
75MskinSkin lesion NOS
76MskinSkin lesion NOS
77FskinSkin lesion NOS
78FskinSkin lesion NOS
79MskinSkin lesion NOS
80FskinTumoral skin lesion
81FskinSkin lesion NOS
82MskinPregressed Cutaneous PTCL
83FskinMycosis fungoides
84MskinSkin lesion NOS
85FskinPlaque
86MskinSkin lesion NOS
87FskinDiffuse pruritis
88FskinSkin lesion NOS
89MskinErythema + axillary adenopathy
90MskinSkin lesion NOS
91MskinSkin lesion NOS
92FskinSkin nodules
93MskinSkin lesion NOS
94MskinPlaque
95FskinSkin lesion NOS
96MskinPapular skin lesions
97FskinSkin lesion NOS
98MskinSkin nodules
99FskinSkin lesion NOS
100MskinSkin lesion NOS
101MskinHIV+, skin lesions, adenopathies
102MskinSuspected follicular mucinosis
103MskinSkin lesion NOS
104MskinErythema and pruritis
105MskinPlaque
106FskinDermatosis
107FskinSkin lesion NOS
108MskinSkin lesion NOS
109FskinSkin lesion NOS
110MskinPlaque

Table A1.

Patients’ characteristics.

NOS = not otherwise specified.

Section and TopicItem #On page #
TITLE/ABSTRACT/KEYWORDS1Identify the article as a study of diagnostic accuracy2
INTRODUCTION2State the research questions or study aims, such as estimating diagnostic accuracy or comparing accuracy between tests or across participant groups.4
METHODS
Participants3Describe the study population: The inclusion and exclusion criteria, setting and locations where the data were collected.5
4Describe participant recruitment: Was recruitment based on presenting symptoms, results from previous tests, or the fact that the participants had received the index tests or the reference standard?Consecutive patients who received the index test (Figure 1)
5Describe participant sampling: Was the study population a consecutive series of participants defined by the selection criteria in items 3 and 4? If not, specify how participants were further selected.Consecutive cases
6Describe data collection: Was data collection planned before the index test and reference standard were performed (prospective study) or after (retrospective study)?Restrospective study
Test methods7Describe the reference standard and its rationale.5
8Describe technical specifications of material and methods involved including how and when measurements were taken, and/or cite references for index tests and reference standard.5-7
9Describe definition of and rationale for the units, cutoffs and/or categories of the results of the index tests and the reference standard.5-7
10Describe the number, training and expertise of the persons executing and reading the index tests and the reference standard.5-6
11Describe whether or not the readers of the index tests and reference standard were blind (masked) to the results of the other test and describe any other clinical information available to the readers.5-7
Statistical methods12Describe methods for calculating or comparing measures of diagnostic accuracy, and the statistical methods used to quantify uncertainty (e.g. 95% confidence intervals).5-6
13Describe methods for calculating test reproducibility, if done.6-7
RESULTS
Participants14Report when study was done, including beginning and ending dates of recruitment.5
15Report clinical and demographic characteristics of the study population (e.g. age, sex, spectrum of presenting symptoms, comorbidity, current treatments, recruitment centers).Table A1
16Report the number of participants satisfying the criteria for inclusion that did or did not undergo the index tests and/or the reference standard; describe why participants failed to receive either test (a flow diagram is strongly recommended).8
Test results17Report time interval from the index tests to the reference standard, and any treatment administered between.6
18Report distribution of severity of disease (define criteria) in those with the target condition; other diagnoses in participants without the target condition.8
19Report a cross tabulation of the results of the index tests (including indeterminate and missing results) by the results of the reference standard; for continuous results, the distribution of the test results by the results of the reference standard.Table A3
20Report any adverse events from performing the index tests or the reference standard./
Estimates21Report estimates of diagnostic accuracy and measures of statistical uncertainty (e.g. 95% confidence intervals).Tables 1 and 2
22Report how indeterminate results, missing responses and outliers of the index tests were handled.6
23Report estimates of variability of diagnostic accuracy between subgroups of participants, readers or centers, if done./
24Report estimates of test reproducibility, if done./
DISCUSSION25Discuss the clinical applicability of the study findings.8-13

Table A2.

STARD requirements for evidence based medicine diagnostic accuracy studies.

Case numberGold standardIndex test: TCRG analysis
1Inflammatory disorderPolyclonal
2MF/SSClonal
3Inflammatory disorderClonal
4Inflammatory disorderPolyclonal
5MF/SSClonal
6Inflammatory disorderClonal
7Inflammatory disorderPolyclonal
8MF/SSClonal
9Inflammatory disorderOligoclonal
11Inflammatory disorderPolyclonal
12non-diagnosticPolyclonal
13Inflammatory disorderPolyclonal
14PTCL/NOSClonal
16B-NHLPolyclonal
17B-NHLClonal
18MF/SSClonal
19non-diagnosticClonal
20PTCL/NOSClonal
21Inflammatory disorderPolyclonal
22non-diagnosticPolyclonal
24Inflammatory disorderClonal
25non-diagnosticClonal
26Inflammatory disorderClonal
27MF/SSClonal
28non-diagnosticClonal
30MF/SSClonal
32B-NHLClonal
33Inflammatory disorderPolyclonal
34Inflammatory disorderPolyclonal
35PTCL/NOSClonal
37MF/SSClonal
38Inflammatory disorderPolyclonal
39MF/SSClonal
40PTCL/NOSClonal
41B-NHLOligoclonal
42PTCL/NOSClonal
43Inflammatory disorderPolyclonal
44Inflammatory disorderPolyclonal
45PTCL/NOSClonal
46T-LGLClonal
47Inflammatory disorderPolyclonal
48Inflammatory disorderPolyclonal
50CTCL/NOSClonal
51Inflammatory disorderPolyclonal
52MF/SSPolyclonal
53PTCL/NOSClonal
54MF/SSClonal
55non-diagnosticPolyclonal
56MF/SSClonal
57Inflammatory disorderPolyclonal
58CTCL/NOSClonal
59non-diagnosticOligoclonal
60PTCL/NOSClonal
61MF/SSPolyclonal
62Inflammatory disorderPolyclonal
64MF/SSClonal
65non-diagnosticPolyclonal
67non-diagnosticPolyclonal
68non-diagnosticOligoclonal
69ALK neg ALCLClonal
70CTCL/NOSClonal
71PTCL/NOSClonal
72non-diagnosticClonal
73Inflammatory disorderPolyclonal
74Inflammatory disorderPolyclonal
75Inflammatory disorderPolyclonal
76MF/SSClonal
77Inflammatory disorderPolyclonal
78Inflammatory disorderPolyclonal
79B-NHLPolyclonal
80CTCL/NOSClonal
81PTCL/NOSPolyclonal
82MF/SSClonal
83Inflammatory disorderPolyclonal
85Inflammatory disorderPolyclonal
86PTCL/NOSPolyclonal
87Inflammatory disorderPolyclonal
88Inflammatory disorderPolyclonal
91Inflammatory disorderPolyclonal
92PTCL/NOSClonal
93B-NHLOligoclonal
94MF/SSClonal
95non-diagnosticOligoclonal
96Inflammatory disorderPolyclonal
98MF/SSClonal
99PTCL/NOSClonal
101PTCL/NOSClonal
102MF/SSClonal
103PTCL/NOSClonal
104Inflammatory disorderPolyclonal
105B-NHLPolyclonal
106Inflammatory disorderPolyclonal
107CTCL/NOSClonal
108Inflammatory disorderPolyclonal
109Inflammatory disorderPolyclonal
110B-NHLPolyclonal

Table A3.

Comparison of Gold standard (histopathological diagnosis) and Index Test (TRG clonality testing).

B-NHL = B-non Hodgkin lymphoma; CTCL = cutaneus T-ceel lymphoma; DLBCL = diffuse large B-cell lymphoma; FL = follicular lymphoma; LGL = large granular lymphocyte leukemia; MF = mycosiss fungoides; MZL = marginal zone lymphoma; NOS = not otherwise specified; PTCL = peripheral T-cell lymphoma; SS = Sezary syndrome.

References

  1. 1. Alaggio R, Amador C, Anagnostopoulos I, et al. The 5th edition of the World Health Organization classification of Haematolymphoid Tumours: Lymphoid neoplasms. Leukemia. 2022;36:1720-1748
  2. 2. Wilcox RA. Cutaneous T-cell lymphoma: 2016 update on diagnosis, risk-stratification, and management. American Journal of Hematology. 2016;91(1):151-165
  3. 3. Weiss LM, Hu E, Wood GS, et al. Clonal rearrangements of T-cell receptor genes in mycosis fungoides and dermatopathic lymphadenopathy. The New England Journal of Medicine. 1985;313(9):539-544
  4. 4. Bertness V, Kirsch I, Hollis G, et al. T-cell receptor gene rearrangements as clinical markers of human T-cell lymphomas. The New England Journal of Medicine. 1985;313(9):534-538
  5. 5. Krangel MS. Mechanics of T cell receptor gene rearrangement. Current Opinion in Immunology. 2009;21(2):133-139
  6. 6. van Dongen JJ, Langerak AW, Bruggemann M, 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(12):2257-2317
  7. 7. Bruggemann M, White H, Gaulard P, et al. Powerful strategy for polymerase chain reaction-based clonality assessment in T-cell malignancies report of the BIOMED-2 concerted action BHM4 CT98-3936. Leukemia. 2007;21(2):215-221
  8. 8. Sandberg Y, Verhaaf B, van Gastel-Mol EJ, et al. Human T-cell lines with well-defined T-cell receptor gene rearrangements as controls for the BIOMED-2 multiplex polymerase chain reaction tubes. Leukemia. 2007;21(2):230-237
  9. 9. Bagg A, Braziel RM, Arber DA, et al. Immunoglobulin heavy chain gene analysis in lymphomas: A multi-center study demonstrating the heterogeneity of performance of polymerase chain reaction assays. The Journal of Molecular Diagnostics. 2002;4(2):81-89
  10. 10. Liu H, Bench AJ, Bacon CM, et al. A practical strategy for the routine use of BIOMED-2 PCR assays for detection of B- and T-cell clonality in diagnostic haematopathology. British Journal of Haematology. 2007;138(1):31-43
  11. 11. Cohen J, Korevaar D, Altman D, et al. STARD 2015 guidelines for reporting diagnostic accuracy studies: Explanation and elaboration. BMJ Open. 2016;6(11):e012799
  12. 12. Zhang B, Beck AH, Taube JM, et al. Combined use of PCR-based TCRG and TCRB clonality tests on paraffin-embedded skin tissue in the differential diagnosis of mycosis fungoides and inflammatory dermatoses. The Journal of Molecular Diagnostics. 2010;12(3):320-327
  13. 13. Lukowsky A, Muche JM, Mobs M, et al. Evaluation of T-cell clonality in archival skin biopsy samples of cutaneous T-cell lymphomas using the biomed-2 PCR protocol. Diagnostic Molecular Pathology. 2010;19(2):70-77
  14. 14. Goeldel AL, Cornillet-Lefebvre P, Durlach A, et al. T-cell receptor gamma gene rearrangement in cutaneous T-cell lymphoma: Comparative study of polymerase chain reaction with denaturing gradient gel electrophoresis and GeneScan analysis. The British Journal of Dermatology. 2009;162(4):822-829
  15. 15. Ponti R, Quaglino P, Novelli M, et al. T-cell receptor gamma gene rearrangement by multiplex polymerase chain reaction/heteroduplex analysis in patients with cutaneous T-cell lymphoma (mycosis fungoides/Sezary syndrome) and benign inflammatory disease: Correlation with clinical, histological and immunophenotypical findings. The British Journal of Dermatology. 2005;153(3):565-573
  16. 16. Hagen MD. Test characteristics. How good is that test? Primary Care. 1995;22(2):213-233
  17. 17. Langerak AW, Molina TJ, Lavender FL, et al. Polymerase chain reaction-based clonality testing in tissue samples with reactive lymphoproliferations: Usefulness and pitfalls. A report of the BIOMED-2 concerted action BMH4-CT98-3936. Leukemia. 2007;21(2):222-229
  18. 18. Schiller PI, Flaig MJ, Puchta U, et al. Detection of clonal T cells in lichen planus. Archives of Dermatological Research. 2000;292(11):568-569
  19. 19. Dereure O, Levi E, Kadin ME. T-cell clonality in pityriasis lichenoides et varioliformis acuta: A heteroduplex analysis of 20 cases. Archives of Dermatology. 2000;136(12):1483-1486
  20. 20. Lukowsky A, Muche JM, Sterry W, et al. Detection of expanded T cell clones in skin biopsy samples of patients with lichen sclerosus et atrophicus by T cell receptor-gamma polymerase chain reaction assays. The Journal of Investigative Dermatology. 2000;115(2):254-259
  21. 21. Meyer N, Mazereeuw-Hautier J, Launay F, et al. Cutaneous T cell lymphoma complicating severe atopic dermatitis. Is making a diagnosis the main challenge? Dermatology. 2009;218(2):168-171
  22. 22. Arana A, Wentworth CE, Fernandez-Vidaurre C, et al. Incidence of cancer in the general population and in patients with or without atopic dermatitis in the U.K. The British Journal of Dermatology. 2010;163(5):1036-1043
  23. 23. Ponti R, Fierro MT, Quaglino P, et al. TCRgamma-chain gene rearrangement by PCR-based GeneScan: Diagnostic accuracy improvement and clonal heterogeneity analysis in multiple cutaneous T-cell lymphoma samples. The Journal of Investigative Dermatology. 2008;128(4):1030-1038
  24. 24. Wood GS, Tung RM, Haeffner AC, et al. Detection of clonal T-cell receptor gamma gene rearrangements in early mycosis fungoides/Sezary syndrome by polymerase chain reaction and denaturing gradient gel electrophoresis (PCR/DGGE). The Journal of Investigative Dermatology. 1994;103(1):34-41
  25. 25. Thurber SE, Zhang B, Kim YH, et al. T-cell clonality analysis in biopsy specimens from two different skin sites shows high specificity in the diagnosis of patients with suggested mycosis fungoides. Journal of the American Academy of Dermatology. 2007;57(5):782-790
  26. 26. Eros N, Karolyi Z, Marschalko M, et al. Clinical, histopathological, immunophenotypic and molecular analysis of 60 patients with cutaneous T-cell infiltrates with follow up of indeterminate cases to identify T-cell lymphoma. Pathology Oncology Research. 2008;14(1):63-67
  27. 27. Foo CC, Tang MB, Chong TK, et al. T-cell receptor-gamma gene analysis in evolving to advancing cutaneous T-cell lymphoma. The Australasian Journal of Dermatology. 2007;48(3):156-160
  28. 28. Vonderheid EC, Bernengo MG, Burg G, et al. Update on erythrodermic cutaneous T-cell lymphoma: Report of the International Society for Cutaneous Lymphomas. Journal of the American Academy of Dermatology. 2002;46(1):95-106
  29. 29. Olsen E, Vonderheid E, Pimpinelli N, et al. Revisions to the staging and classification of mycosis fungoides and Sezary syndrome: A proposal of the International Society for Cutaneous Lymphomas (ISCL) and the cutaneous lymphoma task force of the European Organization of Research and Treatment of cancer (EORTC). Blood. 2007;110(6):1713-1722
  30. 30. Furmanczyk PS, Wolgamot GM, Kussick SJ, et al. Diagnosis of mycosis fungoides with different algorithmic approaches. Journal of Cutaneous Pathology. 2009;37(1):8-14
  31. 31. Ferrara G, Di Blasi A, Zalaudek I, et al. Regarding the algorithm for the diagnosis of early mycosis fungoides proposed by the International Society for Cutaneous Lymphomas: Suggestions from routine histopathology practice. Journal of Cutaneous Pathology. 2008;35(6):549-553
  32. 32. Ralfkiaer U, Hagedorn PH, Bangsgaard N, et al. Diagnostic microRNA profiling in cutaneous T-cell lymphoma (CTCL). Blood. 2011;118(22):5891-5900
  33. 33. Wang L, Ni X, Covington K, et al. Genomic profiling of Sézary syndrome identifies alterations of key T cell signaling and differentiation genes. Nature Genetics. 2015;47(12):12
  34. 34. da Silva AA, Abate F, Khiabanian H, et al. The mutational landscape of cutaneous T cell lymphoma and Sézary syndrome. Nature Genetics. 2015;47(12):5
  35. 35. Choi J, Goh G, Walradt T, et al. Genomic landscape of cutaneous T cell lymphoma. Nature Genetics. 2015;47(9):8
  36. 36. Sufficool K, Lockwood C, Abel H, et al. T-cell clonality assessment by next-generation sequencing improves detection sensitivity in mycosis fungoides. Journal of the American Academy of Dermatology. 2015;73(2):8
  37. 37. Gazzola A, Mannu C, Rossi M, Laginestra MA, Sapienza MR, Fuligni F, et al. The evolution of clonality testing in the diagnosis and monitoring of hematological malignancies. Therapeutic Advances in Hematology. 2014;5:35-47
  38. 38. Greiner TC. Clinical use of next-generation sequencing of trg gene rearrangements has arrived. American Journal of Clinical Pathology. American Society of Clinical Pathologists. 2014;141:302-304
  39. 39. Kohlmann A, Grossmann V, Nadarajah N, Haferlach T. Next-generation sequencing - feasibility and practicality in haematology. British Journal of Haematology. 2013;160:736-753
  40. 40. Walia R, Yeung CCS. An update on molecular biology of cutaneous T cell lymphoma. Frontiers in Oncology. 2020;9:1558. Published 2020 Jan 22. DOI: 10.3389/fonc.2019.01558
  41. 41. Phyo ZH, Shanbhag S, Rozati S. Update on biology of cutaneous T-cell lymphoma. Frontiers in Oncology. 2020;10:765. Published 2020 May 12. DOI: 10.3389/fonc.2020.00765
  42. 42. Weng WK, Armstrong R, Arai S, Desmarais C, Hoppe R, Kim YH. Minimal residual disease monitoring with high-throughput sequencing of T cell receptors in cutaneous T cell lymphoma. Science Translational Medicine. 2013;5(214):214-171

Written By

Fernando Roncolato, Anna Gazzola, Claudia Mannu, Riccardo Donelli, Maryam Etebari, Mohsen Navari and Pier Paolo Piccaluga

Submitted: 18 January 2024 Reviewed: 18 January 2024 Published: 10 April 2024