Is It Time to Assess T Cell Clonality by Next-Generation Sequencing in Mature T Cell Lymphoid Neoplasms? A Scoping Review
Abstract
:1. Introduction
2. Materials and Methods
3. Results
3.1. Review of Publications for Evaluating T Cell Clonality in Patients and Clinical Specimens with Mature Lymphoid Neoplasms and Related Diagnoses by Next-Generation Sequencing
3.1.1. T Cell Receptor Clonality Assessment by Next-Generation Sequencing of the Rearranged DNA of the T Cell Receptor Genes in Patients with Mature T Cell Neoplasms and Related Pathologic Diagnoses
NGS Assays | Assay Manufacturers | TRB Gene Targets | TRG Gene Targets | Target Enrichment |
---|---|---|---|---|
ClonoSEQ assays a | Adaptive Biotechnologies, Seattle, USA | TRB-V(D)J | TRG-VJ | Amplicon-based |
LymphotrackTM assays a | Invivoscribe, San Diego, USA | TRB-V(D)J | TRG-VJ | Amplicon-based |
EuroClonality NGS-amplicon-based assay | EuroClonality group, Europe | TRB-DJ and TRB-V(D)J | TRG-VJ | Amplicon-based |
EuroClonality NGS-capture-based assay (EuroClonality-NDC assay) | EuroClonality group, Europe | TRB-DJ and TRB-V(D)J | TRG-VJ | Capture-based |
3.1.2. Comparative Studies for T Cell Receptor Clonality Assessment by NGS and FL-PCR Assays in Patients with Mature T Cell Neoplasms and Related Pathologic Diagnoses
N Studies (% of 11 Comparative Studies), Total N Patients and Samples Compared in the Studies, and the Sample Types Studied for T Cell Receptor Clonality Assessment by TRG and TRB FL-PCR and NGS Assays | |||||||
---|---|---|---|---|---|---|---|
FL-PCR and NGS Assays | TRG NGS N Cohort Studies and Total N Samples Compared | TRB NGS N Cohort Studies and Total N Samples Compared | EuroClonality–NGS DNA Capture for TRD, TRG, TRB, TRA | Total N Samples Compared | |||
N Studies | Total N Samples | N Studies | Total N Samples | N Cohort Studies | Total N Samples | ||
TRG FL-PCR | N = 5 (45%) Sufficool et al. [59] Kansal et al. [19] Lay et al. [50] Ho et al. [49] Nollet et al. [51] | Total 369 samples in all 5 studies: 95 PB, 68 BM, 117 FFPE, 89 fresh tissues; patients N = 206 in 3 studies [19,51,59], N unavailable [49,50] | N = 2 (18%) Rea et al. [67] Zimmerman et al. [79] | Total 244 skin biopsies and 109 PB, including 100 concurrent PB [67] and 9 PB in SS [79] | TRG and TRB FL-PCR compared with BIOMED-2 FL-PCR assays in 33 mature T cell neoplasms; results given in 7 (21%) of 33 discordant cases [77] | All 33 samples compared with the EuroClonality/BIOMED-2 FL-PCR assays | Total 832 (369 + 110 + 244 + 109) samples studied |
N = 1 (9%), Kirsch et al. [62] | 110 samples from 110 patients a [62] | N = 1 (9%), Kirsch et al. [62] | 110 samples from 110 patients a [62] | ||||
TRB FL-PCR | None | Zero | None | Zero | Zero | ||
EuroClonality/BIOMED-2 TRG and TRB FL-PCR | N = 1 (9%) Bozon et al. [83] | 17 samples (skin) in 7 patients [83] | N = 1 (9%) Gibbs et al. [76] | 102 skin biopsies: 50 FL-PCR tests in 50 patients; 26 NGS tests in 23 patients [76] | N = 1 (9%) Stewart et al. [77] | 33 samples for mature T cell neoplasms; N patients unavailable [77] | 76 (17 + 26 + 33) samples compared by NGS and FL-PCR |
Total N cohort studies and samples | 7 (63.6%) | Total 496 (369 + 110 a + 17) samples in all 7 studies: 95 PB, 68 BM, 117 FFPE, 216 (89 + 110 + 17) fresh tissues | 4 (36.3%) b | Total 489 (244 + 109 +110 a + 26) samples (skin and PB) examined by FL-PCR and NGS | 1 (9%) | 33 samples (24 FFPE, 9 HMW DNA) for mature T cell neoplasms | Total 908 samples studied (22.5% PB, 7.4% BM, 15.5% FFPE, 54.2% fresh tissues) c |
- One (9%) of 11 studies compared TRG FL-PCR with TRG NGS and TRB NGS [62].
- One (9%) of these 11 studies compared the EuroClonality/BIOMED-2 TRG and TRB FL-PCR assays with TRB NGS [76]. The EuroClonality/BIOMED-2 FL-PCR assays analyze TRB-V(D)J, TRB-DJ and TRG-VJ rearrangements.
- One (9%) of these 11 studies compared the EuroClonality/BIOMED-2 TRG and TRB FL-PCR assays with TRG NGS [83].
- One (9%) of 11 studies compared the EuroClonality–NGS DNA Capture assay for evaluating clonality at the TRD, TRG, TRB, and TRA loci with the participating laboratories’ diagnoses based on EuroClonality/BIOMED-2 FL-PCR assays [77].
3.1.3. All 29 Cohort Studies According to the Integrated Clinicopathologic Diagnoses for the Studied Clinical Samples
Cutaneous T Cell Lymphoma
- In 60 patients with mycosis fungoides without peripheral blood involvement, including 50 (83%) with early-stage disease, TRB V(D)J NGS identified dominant clones in peripheral blood in 28% (n = 17) [87]. TRB V(D)J NGS also clarified the presence of discordance in the clone between the skin and blood in 82% (n = 14) of cases [87], which has a better prognosis, including a longer time to systemic treatment than if there are concordant clones present in the skin and blood [71,87].
- Identifying molecular remission in both skin and peripheral blood by TRB V(D)J NGS in patients with advanced mycosis fungoides and Sézary syndrome after an allogeneic hematopoietic stem cell transplant significantly reduced the incidence of progression/relapse versus if NGS detected MRD in either skin or blood [72].
- The absence or low levels of neoplastic T cell receptor sequences by NGS in skin rash associated with the treatment of mycosis fungoides with mogamulizumab, an antibody to CCR4 (chemokine receptor type 4) or after treatment with mechlorethamine gel, helped to distinguish the treatment-associated skin rash from disease progression [75,88].
- NGS identified clonal sequences in specimens obtained from different sites and times [83].
- When analyzed by NGS, peripheral blood samples showed the highest diagnostic specificity of 100% for CTCL, compared with FCI and FL-PCR [76]. Another study found the latter two non-NGS methods to be the least useful for diagnosis and showed 100% diagnostic specificity in skin samples [67]. A French study also showed 100% and 95% diagnostic specificities in skin samples at 25% and 5% tumor cell fraction thresholds, respectively, by NGS [79]. Skin biopsies were often insufficient for analysis by FCI and FL-PCR assays [76].
- Of note, NGS did not help diagnose pediatric pityriasis lichenoides, since a T cell clone may or may not be present in this non-neoplastic disease [81]. This finding further proves that a T cell clone’s presence or absence does not equate to a neoplastic or non-neoplastic disease.
T Cell Prolymphocytic Leukemia
Various Diagnoses of Lymphoid Neoplasms and Non-Neoplastic Diseases
3.1.4. Seven Studies That Used TRG NGS and TRB NGS to Evaluate T Cell Clonality
3.1.5. Two Studies Comparing NGS with Flow Cytometric Immunophenotyping to Evaluate T Cell Clonality
3.1.6. Selected Recent Instructive Case Reports and Cohort Studies That Assessed T Cell Clonality by NGS or FL-PCR
3.2. Additional Publications Relevant to Evaluating T Cell Receptor Clonality by NGS
3.2.1. T Cell Receptor Clonality Assessment by NGS in Patients with Cytopenias
- An NGS study of the T cell repertoire in healthy individuals and patients with cancer showed that a continuous decrease in the diversity of the T cell repertoire in normal individuals begins at the age of 40 years [114]. In untreated patients with hematologic and non-hematologic cancers, there is lower diversity and increased clonality of T cell repertoires. Interestingly, after T cell depleting therapy in patients with cancer, the age-specific repertoire is regained even in older patients [114].
- NGS was used to examine the diversity of the T and B cell repertoires in 25 patients with primary and secondary autoimmune cytopenia who had available follow-up clinical samples. Patients with primary autoimmune cytopenia and 23 patients with active autoimmune hepatitis did not have physiological T cell clusters. These findings suggest that a T cell repertoire signature could act as a biomarker in blood for autoimmune conditions [115].
3.2.2. T Cell Receptor Clonality Evaluation by NGS in Patients with Acute Graft-Versus-Host Disease
3.2.3. T Cell Receptor Clonality Studies by Other Methods, Including Whole Exome, Transcriptome, and Whole Genome Sequencing in Mature T Cell Neoplasms
3.3. Third-Generation Sequencing Compared with Next-Generation Sequencing for T Cell Clonality Assessment
4. Discussion
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Kansal, R. Diagnosis and Molecular Pathology of Lymphoblastic Leukemias and Lymphomas in the Era of Genomics and Precision Medicine: Historical Evolution and Current Concepts—Part 1: Lymphoid Neoplasms. Lymphatics 2023, 1, 55–76. [Google Scholar] [CrossRef]
- Kansal, R. Diagnosis and Molecular Pathology of Lymphoblastic Leukemias and Lymphomas in the Era of Genomics and Precision Medicine: Historical Evolution and Current Concepts—Part 2: B-/T-Cell Acute Lymphoblastic Leukemias. Lymphatics 2023, 1, 118–154. [Google Scholar] [CrossRef]
- Kansal, R. Diagnosis and Molecular Pathology of Lymphoblastic Leukemias and Lymphomas in the Era of Genomics and Precision Medicine: Historical Evolution and Current Concepts—Part 3: Mature Leukemias/Lymphomas. Lymphatics 2023, 1, 155–219. [Google Scholar] [CrossRef]
- Tonegawa, S. Somatic generation of antibody diversity. Nature 1983, 302, 575–581. [Google Scholar] [CrossRef] [PubMed]
- Davis, M.M.; Bjorkman, P.J. T-cell antigen receptor genes and T-cell recognition. Nature 1988, 334, 395–402. [Google Scholar] [CrossRef]
- van Zelm, M.C.; van der Burg, M.; de Ridder, D.; Barendregt, B.H.; de Haas, E.F.; Reinders, M.J.; Lankester, A.C.; Révész, T.; Staal, F.J.; van Dongen, J.J. 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]
- Dik, W.A.; Pike-Overzet, K.; Weerkamp, F.; de Ridder, D.; de Haas, E.F.; Baert, M.R.; van der Spek, P.; Koster, E.E.; Reinders, M.J.; van Dongen, J.J.; 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]
- van Dongen, J.J.; Langerak, A.W.; Brüggemann, M.; Evans, P.A.; 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] [PubMed]
- The Human Genome Organization (HUGO) Nomination Standards. Available online: https://www.hugo-international.org/standards/ (accessed on 24 December 2024).
- Wain, H.M.; Bruford, E.A.; Lovering, R.C.; Lush, M.J.; Wright, M.W.; Povey, S. Guidelines for human gene nomenclature. Genomics 2002, 79, 464–470. [Google Scholar] [CrossRef] [PubMed]
- Bruford, E.A.; Lush, M.J.; Wright, M.W.; Sneddon, T.P.; Povey, S.; Birney, E. The HGNC Database in 2008: A resource for the human genome. Nucleic Acids Res. 2008, 36, D445–D448. [Google Scholar] [CrossRef] [PubMed]
- Lefranc, M.P. Immunoglobulin and T Cell Receptor Genes: IMGT(®) and the Birth and Rise of Immunoinformatics. Front. Immunol. 2014, 5, 22. [Google Scholar] [CrossRef] [PubMed]
- The International Immunogenetics Information System®. IMGT Gene Name Nomenclature for IG and TR of Human and Other Jawed Vertebrates. Available online: https://www.imgt.org/IMGTScientificChart/Nomenclature/IMGTnomenclature.php (accessed on 23 December 2024).
- van Krieken, J.H.; Langerak, A.W.; Macintyre, E.A.; Kneba, M.; Hodges, E.; Sanz, R.G.; Morgan, G.J.; Parreira, A.; Molina, T.J.; Cabeçadas, J.; et al. Improved reliability of lymphoma diagnostics via PCR-based clonality testing: Report of the BIOMED-2 Concerted Action BHM4-CT98-3936. Leukemia 2007, 21, 201–206. [Google Scholar] [CrossRef] [PubMed]
- Evans, P.A.S.; Pott, C.; A Groenen, P.J.T.; Salles, G.; Davi, F.; Berger, F.; Garcia, J.F.; van Krieken, J.H.J.M.; Pals, S.; Kluin, P.; et al. Significantly improved PCR-based clonality testing in B-cell malignancies by use of multiple immunoglobulin gene targets. Report of the BIOMED-2 Concerted Action BHM4-CT98-3936. Leukemia 2007, 21, 207–214. [Google Scholar] [CrossRef] [PubMed]
- Brüggemann, M.; White, H.; Gaulard, P.; Garcia-Sanz, R.; Gameiro, P.; Oeschger, S.; Jasani, B.; Ott, M.; Delsol, G.; Orfao, A.; 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, 215–221. [Google Scholar] [CrossRef] [PubMed]
- Langerak, A.W.; Groenen, P.J.; Brüggemann, M.; Beldjord, K.; Bellan, C.; Bonello, L.; Boone, E.; Carter, G.I.; Catherwood, M.; Davi, F.; et al. EuroClonality/BIOMED-2 guidelines for interpretation and reporting of Ig/TCR clonality testing in suspected lymphoproliferations. Leukemia 2012, 26, 2159–2171. [Google Scholar] [CrossRef]
- Armand, M.; Derrieux, C.; Beldjord, K.; Wabeke, T.; Lenze, D.; Boone, E.; Bruggemann, M.; Evans, P.A.S.; Gameiro, P.; Hummel, M.; et al. A New and Simple TRG Multiplex PCR Assay for Assessment of T-cell Clonality: A Comparative Study from the EuroClonality Consortium. Hemasphere 2019, 3, e255. [Google Scholar] [CrossRef] [PubMed]
- Kansal, R.; Grody, W.W.; Zhou, J.; Dong, L.; Li, X. The Value of T-Cell Receptor γ (TRG) Clonality Evaluation by Next-Generation Sequencing in Clinical Hematolymphoid Tissues. Am. J. Clin. Pathol. 2018, 150, 193–223. [Google Scholar] [CrossRef] [PubMed]
- Langerak, A.W.; van Krieken, J.H.; Wolvers-Tettero, I.L.; Kerkhof, E.; Mulder, A.H.; Vrints, L.W.; Coebergh, J.W.; Schuuring, E.; Kluin, P.M.; van Dongen, J.J. The role of molecular analysis of immunoglobulin and T cell receptor gene rearrangements in the diagnosis of lymphoproliferative disorders. J. Clin. Pathol. 2001, 54, 565–567. [Google Scholar] [CrossRef] [PubMed]
- Gazzola, A.; Mannu, C.; Rossi, M.; Laginestra, M.A.; Sapienza, M.R.; Fuligni, F.; Etebari, M.; Melle, F.; Sabattini, E.; Agostinelli, C.; et al. The evolution of clonality testing in the diagnosis and monitoring of hematological malignancies. Ther. Adv. Hematol. 2014, 5, 35–47. [Google Scholar] [CrossRef] [PubMed]
- Alaggio, R.; Amador, C.; Anagnostopoulos, I.; Attygalle, A.D.; Araujo, I.B.O.; Berti, E.; Bhagat, G.; Borges, A.M.; Boyer, D.; Calaminici, M.; et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Lymphoid Neoplasms. Leukemia 2022, 36, 1720–1748. [Google Scholar] [CrossRef] [PubMed]
- Khoury, J.D.; Solary, E.; Abla, O.; Akkari, Y.; Alaggio, R.; Apperley, J.F.; Bejar, R.; Berti, E.; Busque, L.; Chan, J.K.C.; et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Myeloid and Histiocytic/Dendritic Neoplasms. Leukemia 2022, 36, 1703–1719. [Google Scholar] [CrossRef] [PubMed]
- Arber, D.A.; Orazi, A.; Hasserjian, R.P.; Borowitz, M.J.; Calvo, K.R.; Kvasnicka, H.M.; Wang, S.A.; Bagg, A.; Barbui, T.; Branford, S.; et al. International Consensus Classification of Myeloid Neoplasms and Acute Leukemias: Integrating morphologic, clinical, and genomic data. Blood 2022, 140, 1200–1228. [Google Scholar] [CrossRef] [PubMed]
- Campo, E.; Jaffe, E.S.; Cook, J.R.; Quintanilla-Martinez, L.; Swerdlow, S.H.; Anderson, K.C.; Brousset, P.; Cerroni, L.; de Leval, L.; Dirnhofer, S.; et al. The International Consensus Classification of Mature Lymphoid Neoplasms: A report from the Clinical Advisory Committee. Blood 2022, 140, 1229–1253. [Google Scholar] [CrossRef] [PubMed]
- Kansal, R. The World Health Organization (WHO) Classification of Tumors with Emphasis on the Classification of Hematolymphoid Neoplasms. In Precision Medicine: Where Are We and Where Are We Going? Kansal, R., Ed.; Nova Science Publishers, Inc.: Hauppage, NY, USA, 2023; pp. 315–416. [Google Scholar] [CrossRef]
- Naresh, K.N.; Medeiros, L.J.; WHO Fifth Edition Classification Project. Introduction to the Fifth Edition of the World Health Organization Classification of Tumors of Hematopoietic and Lymphoid Tissues. Mod. Pathol. 2023, 36, 100330. [Google Scholar] [CrossRef] [PubMed]
- Miranda, R.N.; Amador, C.; Chan, J.K.C.; Guitart, J.; Rech, K.L.; Medeiros, L.J.; Naresh, K.N.; WHO Fifth Edition Classification Project. Fifth Edition of the World Health Organization Classification of Tumors of the Hematopoietic and Lymphoid Tissues: Mature T-Cell, NK-Cell, and Stroma-Derived Neoplasms of Lymphoid Tissues. Mod. Pathol. 2024, 37, 100512. [Google Scholar] [CrossRef] [PubMed]
- Bhat, P.V.; Jakobiec, F.A.; Papaliodis, G.; Sobrin, L. Primary T-cell lymphoma of the retina and cerebellum: Immunophenotypic and gene rearrangement confirmation. Am. J. Ophthalmol. 2009, 148, 350–360. [Google Scholar] [CrossRef] [PubMed]
- Civallero, M.; Schroers-Martin, J.G.; Horwitz, S.; Manni, M.; Stepanishyna, Y.; Cabrera, M.E.; Vose, J.; Spina, M.; Hitz, F.; Nagler, A.; et al. Long-term outcome of peripheral T-cell lymphomas: Ten-year follow-up of the International Prospective T-cell Project. Br. J. Haematol. 2024, 205, 166–174. [Google Scholar] [CrossRef] [PubMed]
- Vose, J.; Armitage, J.; Weisenburger, D.; International T-Cell Lymphoma Project. International peripheral T-cell and natural killer/T-cell lymphoma study: Pathology findings and clinical outcomes. J. Clin. Oncol. 2008, 26, 4124–4130. [Google Scholar] [CrossRef] [PubMed]
- Hristov, A.C.; Tejasvi, T.; Wilcox, R.A. Cutaneous T-cell lymphomas: 2023 update on diagnosis, risk-stratification, and management. Am. J. Hematol. 2023, 98, 193–209. [Google Scholar] [CrossRef] [PubMed]
- Willemze, R.; Cerroni, L.; Kempf, W.; Berti, E.; Facchetti, F.; Swerdlow, S.H.; Jaffe, E.S. The 2018 update of the WHO-EORTC classification for primary cutaneous lymphomas. Blood 2019, 133, 1703–1714. [Google Scholar] [CrossRef] [PubMed]
- Kempf, W.; Mitteldorf, C.; Cerroni, L.; Willemze, R.; Berti, E.; Guenova, E.; Scarisbrick, J.J.; Battistella, M. Classifications of cutaneous lymphomas and lymphoproliferative disorders: An update from the EORTC cutaneous lymphoma histopathology group. J. Eur. Acad. Dermatol. Venereol. 2024, 38, 1491–1503. [Google Scholar] [CrossRef] [PubMed]
- Böck, J.; Maurus, K.; Gerhard-Hartmann, E.; Brändlein, S.; Kurz, K.S.; Ott, G.; Anagnostopoulos, I.; Rosenwald, A.; Zamò, A. Targeted panel sequencing in the routine diagnosis of mature T- and NK-cell lymphomas: Report of 128 cases from two German reference centers. Front. Oncol. 2023, 13, 1231601. [Google Scholar] [CrossRef] [PubMed]
- Szczepański, T.; Beishuizen, A.; Pongers-Willemse, M.J.; Hählen, K.; Van Wering, E.R.; Wijkhuijs, A.J.; Tibbe, G.J.; De Bruijn, M.A.; Van Dongen, J.J. 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] [PubMed]
- Groenen, P.J.; Langerak, A.W.; van Dongen, J.J.; van Krieken, J.H. Pitfalls in TCR gene clonality testing: Teaching cases. J. Hematop. 2008, 1, 97–109. [Google Scholar] [CrossRef] [PubMed]
- Langerak, A.W.; Brüggemann, M.; Davi, F.; Darzentas, N.; van Dongen, J.J.M.; Gonzalez, D.; Cazzaniga, G.; Giudicelli, V.; Lefranc, M.P.; Giraud, M.; et al. High-Throughput Immunogenetics for Clinical and Research Applications in Immunohematology: Potential and Challenges. J. Immunol. 2017, 198, 3765–3774. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Scheijen, B.; Meijers, R.W.J.; Rijntjes, J.; van der Klift, M.Y.; Möbs, M.; Steinhilber, J.; Reigl, T.; van den Brand, M.; Kotrová, M.; Ritter, J.M.; et al. Next-generation sequencing of immunoglobulin gene rearrangements for clonality assessment: A technical feasibility study by EuroClonality-NGS. Leukemia 2019, 33, 2227–2240. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Bystry, V.; Reigl, T.; Krejci, A.; Demko, M.; Hanakova, B.; Grioni, A.; Knecht, H.; Schlitt, M.; Dreger, P.; Sellner, L.; et al. ARResT/Interrogate: An interactive immunoprofiler for IG/TR NGS data. Bioinformatics 2017, 33, 435–437. [Google Scholar] [CrossRef] [PubMed]
- van den Brand, M.; Rijntjes, J.; Möbs, M.; Steinhilber, J.; van der Klift, M.Y.; Heezen, K.C.; Kroeze, L.I.; Reigl, T.; Porc, J.; Darzentas, N.; et al. Next-Generation Sequencing-Based Clonality Assessment of Ig Gene Rearrangements: A Multicenter Validation Study by EuroClonality-NGS. J. Mol. Diagn. 2021, 23, 1105–1115. [Google Scholar] [CrossRef] [PubMed]
- van Bladel, D.A.G.; van der Last-Kempkes, J.L.M.; Scheijen, B.; Groenen, P.J.T.A.; EuroClonality Consortium. Next-Generation Sequencing-Based Clonality Detection of Immunoglobulin Gene Rearrangements in B-Cell Lymphoma. Methods Mol. Biol. 2022, 2453, 7–42. [Google Scholar] [CrossRef] [PubMed]
- van den Brand, M.; Möbs, M.; Otto, F.; Kroeze, L.I.; Gonzalez de Castro, D.; Stamatopoulos, K.; Davi, F.; Bravetti, C.; Kolijn, P.M.; Vlachonikola, E.; et al. EuroClonality-NGS Recommendations for Evaluation of B-Cell Clonality Analysis by Next-Generation Sequencing: A Structured Approach with the DEPART Algorithm. J. Mol. Diagn. 2023, 25, 729–739. [Google Scholar] [CrossRef] [PubMed]
- van Bladel, D.A.G.; Stevens, W.B.C.; Kroeze, L.I.; de Groen, R.A.L.; de Groot, F.A.; van der Last-Kempkes, J.L.M.; Berendsen, M.R.; Rijntjes, J.; Luijks, J.A.C.W.; Bonzheim, I.; et al. A significant proportion of classic Hodgkin lymphoma recurrences represents clonally unrelated second primary lymphoma. Blood Adv. 2023, 7, 5911–5924. [Google Scholar] [CrossRef] [PubMed]
- Berendsen, M.R.; van Bladel, D.A.G.; Hesius, E.; Berganza Irusquieta, C.; Rijntjes, J.; van Spriel, A.B.; van der Spek, E.; Pruijt, J.F.M.; Kroeze, L.I.; Hebeda, K.M.; et al. Clonal Relationship and Mutation Analysis in Lymphoplasmacytic Lymphoma/Waldenström Macroglobulinemia Associated With Diffuse Large B-cell Lymphoma. Hemasphere 2023, 7, e976. [Google Scholar] [CrossRef] [PubMed]
- Berendsen, M.R.; Bladel, D.A.G.V.; Hesius, E.; de Groot, F.A.; Kroeze, L.I.; Rijntjes, J.; Luijks, J.A.C.W.; Hoevenaars, B.; Halilovic, A.; Nooijen, P.; et al. Detection of Second Primary Lymphoma in Late Diffuse Large B-cell Lymphoma Recurrences. Mod. Pathol. 2023, 36, 100119. [Google Scholar] [CrossRef] [PubMed]
- Ho, C.C.; Tung, J.K.; Zehnder, J.L.; Zhang, B.M. Validation of a Next-Generation Sequencing-Based T-Cell Receptor Gamma Gene Rearrangement Diagnostic Assay: Transitioning from Capillary Electrophoresis to Next-Generation Sequencing. J. Mol. Diagn. 2021, 23, 805–815. [Google Scholar] [CrossRef] [PubMed]
- Lay, L.; Stroup, B.; Payton, J.E. Validation and interpretation of IGH and TCR clonality testing by Ion Torrent S5 NGS for diagnosis and disease monitoring in B and T cell cancers. Pract. Lab. Med. 2020, 22, e00191. [Google Scholar] [CrossRef] [PubMed]
- Nollet, F.; Vanhouteghem, K.; Vermeire, S.; Maelbrancke, E.; Emmerechts, J.; Devos, H.; Cauwelier, B. Evaluation of next-generation sequencing-based clonality analysis of T-cell receptor gamma gene rearrangements based on a new interpretation algorithm. Int. J. Lab. Hematol. 2019, 41, 242–249. [Google Scholar] [CrossRef] [PubMed]
- Arcila, M.E.; Yu, W.; Syed, M.; Kim, H.; Maciag, L.; Yao, J.; Ho, C.; Petrova, K.; Moung, C.; Salazar, P.; et al. Establishment of Immunoglobulin Heavy (IGH) Chain Clonality Testing by Next-Generation Sequencing for Routine Characterization of B-Cell and Plasma Cell Neoplasms. J. Mol. Diagn. 2019, 21, 330–342. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Ho, C.; Yu, W.; Huang, Y.; Miller, J.; Gao, Q.; Syed, M.; Ma, Y.; Wang, M.; Maciag, L.; et al. Quantification of Measurable Residual Disease Detection by Next-Generation Sequencing-Based Clonality Testing in B-Cell and Plasma Cell Neoplasms. J. Mol. Diagn. 2024, 26, 168–178. [Google Scholar] [CrossRef] [PubMed]
- Ma, S.B.; Lin, W.; Campbell, J.; Clerici, K.; White, D.; Yeung, D.; Gorniak, M.; Fleming, S.; Fong, C.Y.; Agarwal, R. Laboratory validation and clinical utility of next-generation sequencing-based IGH/TCR clonality testing for the monitoring of measurable residual disease in acute lymphoblastic leukaemia: Real-world experience at Austin Pathology. Pathology 2024, 56, 982–992. [Google Scholar] [CrossRef] [PubMed]
- Tricco, A.C.; Lillie, E.; Zarin, W.; O’Brien, K.K.; Colquhoun, H.; Levac, D.; Moher, D.; Peters, M.D.J.; Horsley, T.; Weeks, L.; et al. PRISMA Extension for Scoping Reviews (PRISMA-ScR): Checklist and Explanation. Ann. Intern. Med. 2018, 169, 467–473. [Google Scholar] [CrossRef] [PubMed]
- Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef] [PubMed]
- Robins, H.S.; Campregher, P.V.; Srivastava, S.K.; Wacher, A.; Turtle, C.J.; Kahsai, O.; Riddell, S.R.; Warren, E.H.; Carlson, C.S. Comprehensive assessment of T-cell receptor beta-chain diversity in alphabeta T cells. Blood 2009, 114, 4099–4107. [Google Scholar] [CrossRef] [PubMed]
- Weng, W.K.; Armstrong, R.; Arai, S.; Desmarais, C.; Hoppe, R.; Kim, Y.H. Minimal residual disease monitoring with high-throughput sequencing of T cell receptors in cutaneous T cell lymphoma. Sci. Transl. Med. 2013, 5, 214ra171. [Google Scholar] [CrossRef] [PubMed]
- Sufficool, K.E.; Lockwood, C.M.; Abel, H.J.; Hagemann, I.S.; Schumacher, J.A.; Kelley, T.W.; Duncavage, E.J. T-cell clonality assessment by next-generation sequencing improves detection sensitivity in mycosis fungoides. J. Am. Acad. Dermatol. 2015, 73, 228–236.e2. [Google Scholar] [CrossRef] [PubMed]
- Willemze, R.; Jaffe, E.S.; Burg, G.; Cerroni, L.; Berti, E.; Swerdlow, S.H.; Ralfkiaer, E.; Chimenti, S.; Diaz-Perez, J.L.; Duncan, L.M.; et al. WHO-EORTC classification for cutaneous lymphomas. Blood 2005, 105, 3768–3785. [Google Scholar] [CrossRef] [PubMed]
- Carlson, C.S.; Emerson, R.O.; Sherwood, A.M.; Desmarais, C.; Chung, M.W.; Parsons, J.M.; Steen, M.S.; LaMadrid-Herrmannsfeldt, M.A.; Williamson, D.W.; Livingston, R.J.; et al. Using synthetic templates to design an unbiased multiplex PCR assay. Nat. Commun. 2013, 4, 2680. [Google Scholar] [CrossRef] [PubMed]
- Kirsch, I.R.; Watanabe, R.; O’Malley, J.T.; Williamson, D.W.; Scott, L.L.; Elco, C.P.; Teague, J.E.; Gehad, A.; Lowry, E.L.; LeBoeuf, N.R.; et al. TCR sequencing facilitates diagnosis and identifies mature T cells as the cell of origin in CTCL. Sci. Transl. Med. 2015, 7, 308ra158. [Google Scholar] [CrossRef] [PubMed]
- van der Velden, V.H.; 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] [PubMed]
- Sellner, L.; Brüggemann, M.; Schlitt, M.; Knecht, H.; Herrmann, D.; Reigl, T.; Krejci, A.; Bystry, V.; Darzentas, N.; Rieger, M.; et al. GvL effects in T-prolymphocytic leukemia: Evidence from MRD kinetics and TCR repertoire analyses. Bone Marrow Transplant. 2017, 52, 544–551, Erratum in Bone Marrow Transplant 2017, 52, 656. [Google Scholar] [CrossRef]
- de Masson, A.; O’Malley, J.T.; Elco, C.P.; Garcia, S.S.; Divito, S.J.; Lowry, E.L.; Tawa, M.; Fisher, D.C.; Devlin, P.M.; Teague, J.E.; et al. High-throughput sequencing of the T cell receptor β gene identifies aggressive early-stage mycosis fungoides. Sci. Transl. Med. 2018, 10, eaar5894. [Google Scholar] [CrossRef] [PubMed]
- Cushman-Vokoun, A.M.; Connealy, S.; Greiner, T.C. Assay design affects the interpretation of T-cell receptor gamma gene rearrangements: Comparison of the performance of a one-tube assay with the BIOMED-2-based TCRG gene clonality assay. J. Mol. Diagn. 2010, 12, 787–796. [Google Scholar] [CrossRef] [PubMed]
- Rea, B.; Haun, P.; Emerson, R.; Vignali, M.; Farooqi, M.; Samimi, S.; Elenitsas, R.; Kirsch, I.; Bagg, A. Role of high-throughput sequencing in the diagnosis of cutaneous T-cell lymphoma. J. Clin. Pathol. 2018, 71, 814–820. [Google Scholar] [CrossRef] [PubMed]
- van den Beemd, R.; Boor, P.P.; van Lochem, E.G.; Hop, W.C.; Langerak, A.W.; Wolvers-Tettero, I.L.; Hooijkaas, H.; van Dongen, J.J. Flow cytometric analysis of the Vbeta repertoire in healthy controls. Cytometry 2000, 40, 336–345. [Google Scholar] [CrossRef] [PubMed]
- Langerak, A.W.; van Den Beemd, R.; Wolvers-Tettero, I.L.; Boor, P.P.; van Lochem, E.G.; Hooijkaas, H.; van Dongen, J.J. Molecular and flow cytometric analysis of the Vbeta repertoire for clonality assessment in mature TCR alphabeta T-cell proliferations. Blood 2001, 98, 165–173. [Google Scholar] [CrossRef] [PubMed]
- Kotrova, M.; Novakova, M.; Oberbeck, S.; Mayer, P.; Schrader, A.; Knecht, H.; Hrusak, O.; Herling, M.; Brüggemann, M. Next-generation amplicon TRB locus sequencing can overcome limitations of flow-cytometric Vβ expression analysis and confirms clonality in all T-cell prolymphocytic leukemia cases. Cytometry A 2018, 93, 1118–1124. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Rea, B.; Haun, P.; Emerson, R.; Kirsch, I.; Bagg, A. High-throughput sequencing of the T-cell receptor β chain gene distinguishes 2 subgroups of cutaneous T-cell lymphoma. J. Am. Acad. Dermatol. 2019, 80, 1148–1150.e1. [Google Scholar] [CrossRef] [PubMed]
- Weng, W.K.; Arai, S.; Rezvani, A.; Johnston, L.; Lowsky, R.; Miklos, D.; Shizuru, J.; Muffly, L.; Meyer, E.; Negrin, R.S.; et al. Nonmyeloablative allogeneic transplantation achieves clinical and molecular remission in cutaneous T-cell lymphoma. Blood Adv. 2020, 4, 4474–4482. [Google Scholar] [CrossRef] [PubMed]
- U.S. Food and Drug Administration. FDA Approves Mogamulizumab-Kpkc for Mycosis Fungoides or SÉZARY Syndrome. 2018. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-mogamulizumab-kpkc-mycosis-fungoides-or-sezary-syndrome (accessed on 12 September 2024).
- Zhang, B.; Beck, A.H.; Taube, J.M.; Kohler, S.; Seo, K.; Zwerner, J.; Viakhereva, N.; Sundram, U.; Kim, Y.H.; Schrijver, I.; 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. J. Mol. Diagn. 2010, 12, 320–327. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.Y.; Hirotsu, K.E.; Neal, T.M.; Raghavan, S.S.; Kwong, B.Y.; Khodadoust, M.S.; Brown, R.A.; Novoa, R.A.; Kim, Y.H.; Rieger, K.E. Histopathologic Characterization of Mogamulizumab-associated Rash. Am. J. Surg. Pathol. 2020, 44, 1666–1676. [Google Scholar] [CrossRef] [PubMed]
- Gibbs, J.D.; Ma, S.; Kim, A.; Seminario-Vidal, L.; Sokol, L.; Zhang, H.; Zhang, X.; Sagatys, E.; Chen, P.L.; Messina, J.L. Utility of flow cytometry and gene rearrangement analysis in tissue and blood of patients with suspected cutaneous T-cell lymphoma. Oncol. Rep. 2021, 45, 349–358. [Google Scholar] [CrossRef] [PubMed]
- Stewart, J.P.; Gazdova, J.; Darzentas, N.; Wren, D.; Proszek, P.; Fazio, G.; Songia, S.; Alcoceba, M.; Sarasquete, M.E.; Villarese, P.; et al. Validation of the EuroClonality-NGS DNA capture panel as an integrated genomic tool for lymphoproliferative disorders. Blood Adv. 2021, 5, 3188–3198. [Google Scholar] [PubMed]
- Olsen, E.A.; Whittaker, S.; Kim, Y.H.; Duvic, M.; Prince, H.M.; Lessin, S.R.; Wood, G.S.; Willemze, R.; Demierre, M.F.; Pimpinelli, N.; et al. Clinical end points and response criteria in mycosis fungoides and Sézary syndrome: A consensus statement of the International Society for Cutaneous Lymphomas, the United States Cutaneous Lymphoma Consortium, and the Cutaneous Lymphoma Task Force of the European Organisation for Research and Treatment of Cancer. J. Clin. Oncol. 2011, 29, 2598–2607. [Google Scholar] [CrossRef] [PubMed]
- Zimmermann, C.; Boisson, M.; Ram-Wolff, C.; Sadoux, A.; Louveau, B.; Vignon-Pennamen, M.D.; Rivet, J.; Cayuela, J.M.; Dobos, G.; Moins-Teisserenc, H.; et al. Diagnostic performance of high-throughput sequencing of the T-cell receptor beta gene for the diagnosis of cutaneous T-cell lymphoma. Br. J. Dermatol. 2021, 185, 679–680. [Google Scholar] [CrossRef] [PubMed]
- Trum, N.A.; Zain, J.; Martinez, X.U.; Parekh, V.; Afkhami, M.; Abdulla, F.; Carson, K.R.; Rosen, S.T.; Bennett, C.L.; Querfeld, C. Mogamulizumab efficacy is underscored by its associated rash that mimics cutaneous T-cell lymphoma: A retrospective single-centre case series. Br. J. Dermatol. 2022, 186, 153–166. [Google Scholar] [CrossRef] [PubMed]
- Raghavan, S.S.; Wang, J.Y.; Gru, A.A.; Marqueling, A.L.; Teng, J.M.C.; Brown, R.A.; Novoa, R.A.; Kim, Y.; Zehnder, J.; Zhang, B.M.; et al. Next-generation sequencing confirms T-cell clonality in a subset of pediatric pityriasis lichenoides. J. Cutan. Pathol. 2022, 49, 252–260. [Google Scholar] [CrossRef] [PubMed]
- van Santen, S.; Zoutman, W.H.; de Masson, A.; Quint, K.D.; Willemze, R.; Gerard, N.; Teague, J.E.; Kupper, T.S.; Clark, R.A.; Tensen, C.P.; et al. Tumor Clone Frequency Calculation Using High-Throughput Sequencing of the TCRβ Gene in Patients with Folliculotropic Mycosis Fungoides. J. Investig. Dermatol. 2022, 142, 2544–2546.e2. [Google Scholar] [CrossRef] [PubMed]
- Bozon, A.; Avinens, O.; Lacheretz-Szablewski, V.; Dereure, O.; Thévenin, C. Spatial and temporal homogeneity of T-cell receptor gamma chain rearrangements in mycosis fungoides: A next-generation sequencing analysis. J. Am. Acad. Dermatol. 2022, 87, 908–910. [Google Scholar] [CrossRef] [PubMed]
- Tung, J.K.; Jangam, D.; Ho, C.C.; Fung, E.; Khodadoust, M.S.; Kim, Y.H.; Zehnder, J.L.; Stehr, H.; Zhang, B.M. Minimal/Measurable Residual Disease Monitoring in Patients with Lymphoid Neoplasms by High-Throughput Sequencing of the T-Cell Receptor. J. Mol. Diagn. 2023, 25, 331–341. [Google Scholar] [CrossRef] [PubMed]
- Gleason, L.; Cohen, A.; South, A.P.; Porcu, P.; Nikbakht, N. Emergence of Malignant T-Cell Intraclonal CDR3 Variants in Mycosis Fungoides. JAMA Dermatol. 2023, 159, 888–890. [Google Scholar] [CrossRef] [PubMed]
- Olsen, E.A.; Whittaker, S.; Willemze, R.; Pinter-Brown, L.; Foss, F.; Geskin, L.; Schwartz, L.; Horwitz, S.; Guitart, J.; Zic, J.; et al. Primary cutaneous lymphoma: Recommendations for clinical trial design and staging update from the ISCL, USCLC, and EORTC. Blood 2022, 140, 419–437. [Google Scholar] [CrossRef] [PubMed]
- Joffe, D.; Bhatti, S.; Banner, L.; Zaya, R.; Gleason, L.; Mishra, A.; Kirsch, I.; Porcu, P.; Nikbakht, N. The overlap of skin and blood T-cell clones in early-stage mycosis fungoides. Blood Adv. 2023, 7, 6206–6210. [Google Scholar] [CrossRef] [PubMed]
- Bhatti, S.; Joffe, D.; Banner, L.; Talasila, S.; Mandel, J.; Lee, J.; Porcu, P.; Nikbakht, N. Utility of T-cell immunosequencing in distinguishing mycosis fungoides progression from treatment related cutaneous adverse events. Front. Med. 2023, 10, 1243459. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, P.C.; Nguyen, T.; Wilson, C.; Tiong, I.S.; Baldwin, K.; Nguyen, V.; Came, N.; Blombery, P.; Westerman, D.A. Evaluation of T-cell clonality by anti-TRBC1 antibody-based flow cytometry and correlation with T-cell receptor sequencing. Br. J. Haematol. 2024, 204, 910–920. [Google Scholar] [CrossRef] [PubMed]
- Kalinova, M.; Mrhalova, M.; Kabickova, E.; Svaton, M.; Skotnicova, A.; Prouzova, Z.; Krenova, Z.; Kolenova, A.; Divoka, M.; Fronkova, E.; et al. Molecular Screening in Anaplastic Lymphoma Kinase-Positive Anaplastic Large Cell Lymphoma: Anaplastic Lymphoma Kinase Analysis, Next-Generation Sequencing Fusion Gene Detection, and T-Cell Receptor Immunoprofiling. Mod. Pathol. 2024, 37, 100428. [Google Scholar] [CrossRef] [PubMed]
- Thermofisher Scientific Oncomine TCR Beta-SR Assay. Available online: https://www.thermofisher.com/us/en/home/clinical/preclinical-companion-diagnostic-development/oncomine-tcr-beta-sr-assay.html (accessed on 4 December 2024).
- Harland, L.; Borgmann, V.; Otto, F.; Overkamp, M.; Bonzheim, I.; Fend, F.; Quintanilla-Martinez, L.; Nann, D. Clonal Hematopoiesis and Bone Marrow Infiltration in Patients With Follicular Helper T-Cell Lymphoma of Angioimmunoblastic Type. Mod. Pathol. 2024, 37, 100519. [Google Scholar] [CrossRef] [PubMed]
- Shinohara, M.M.; Rieger, K.E.; Sundram, U.; Fung, M.A.; Hristov, A.C. Assessing T-cell receptor clonality by next-generation sequencing in atypical cutaneous lymphoid infiltrates and cutaneous T-cell lymphoma: A scoping review. J. Cutan. Pathol. 2024, 51, 813–819. [Google Scholar] [CrossRef] [PubMed]
- Vidal, C.I.; Armbrect, E.A.; Andea, A.A.; Bohlke, A.K.; Comfere, N.I.; Hughes, S.R.; Kim, J.; Kozel, J.A.; Lee, J.B.; Linos, K.; et al. Appropriate use criteria in dermatopathology: Initial recommendations from the American Society of Dermatopathology. J. Cutan. Pathol. 2018, 45, 563–580. [Google Scholar] [CrossRef] [PubMed]
- Sandberg, Y.; van Gastel-Mol, E.J.; Verhaaf, B.; Lam, K.H.; van Dongen, J.J.; Langerak, A.W. BIOMED-2 multiplex immunoglobulin/T-cell receptor polymerase chain reaction protocols can reliably replace Southern blot analysis in routine clonality diagnostics. J. Mol. Diagn. 2005, 7, 495–503. [Google Scholar] [CrossRef] [PubMed]
- Comfere, N.; Sundram, U.; Hurley, M.Y.; Swick, B. Views of dermatopathologists about clonality assays in the diagnosis of cutaneous T-cell and B-cell lymphoproliferative disorders. J. Cutan. Pathol. 2018, 45, 39–47. [Google Scholar] [CrossRef] [PubMed]
- Giudicelli, V.; Brochet, X.; Lefranc, M.P. IMGT/V-QUEST: IMGT standardized analysis of the immunoglobulin (IG) and T cell receptor (TR) nucleotide sequences. Cold Spring Harb. Protoc. 2011, 2011, 695–715. [Google Scholar] [CrossRef] [PubMed]
- Miller, S.; Vandergriff, T.; Woodworth Goff, H.; Xu, J.; Oliver, D. Comparison of 2 T-Cell Receptor-γ Clonality Assays on Skin Biopsies Suspicious for Mycosis Fungoides. Am. J. Dermatopathol. 2024, 46, 581–587. [Google Scholar] [CrossRef] [PubMed]
- Ibrahim, H.A.; Menasce, L.P.; Pomplun, S.; Burke, M.; Bower, M.; Naresh, K.N. Presence of monoclonal T-cell populations in B-cell post-transplant lymphoproliferative disorders. Mod. Pathol. 2011, 24, 232–240. [Google Scholar] [CrossRef] [PubMed]
- Willemze, R. Cutaneous lymphoproliferative disorders: Back to the future. J. Cutan. Pathol. 2024, 51, 468–476. [Google Scholar] [CrossRef] [PubMed]
- Maciocia, P.M.; Wawrzyniecka, P.A.; Philip, B.; Ricciardelli, I.; Akarca, A.U.; Onuoha, S.C.; Legut, M.; Cole, D.K.; Sewell, A.K.; Gritti, G.; et al. Targeting the T cell receptor β-chain constant region for immunotherapy of T cell malignancies. Nat. Med. 2017, 23, 1416–1423. [Google Scholar] [CrossRef] [PubMed]
- Tunnacliffe, A.; Kefford, R.; Milstein, C.; Forster, A.; Rabbitts, T.H. Sequence and evolution of the human T-cell antigen receptor beta-chain genes. Proc. Natl. Acad. Sci. USA 1985, 82, 5068–5072. [Google Scholar] [CrossRef] [PubMed]
- Hwang, J.K.; Wang, E.; Neff, J.L.; Wang, J. A Cautionary Tale: Florid Splenic γδ T-cell Proliferation and False-Positive T-cell Clonality by PCR Leads to a Grave Misdiagnosis. Clin. Lymphoma Myeloma Leuk. 2021, 21, e748–e751. [Google Scholar] [CrossRef] [PubMed]
- Foss, F.M.; Horwitz, S.M.; Civallero, M.; Bellei, M.; Marcheselli, L.; Kim, W.S.; Cabrera, M.E.; Dlouhy, I.; Nagler, A.; Advani, R.H.; et al. Incidence and outcomes of rare T cell lymphomas from the T Cell Project: Hepatosplenic, enteropathy associated and peripheral gamma delta T cell lymphomas. Am. J. Hematol. 2020, 95, 151–155. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.Y.; Collins, G.P.; Soilleux, E.; Eyre, T.A. Acute EBV masquerading as peripheral T-cell lymphoma. BMJ Case Rep. 2016, 2016, bcr2015213573. [Google Scholar] [CrossRef] [PubMed]
- Rojansky, R.; Fernandez-Pol, S.; Wang, E.; Rieger, K.E.; Novoa, R.A.; Zehnder, J.L.; Kunder, C.A.; Kim, Y.H.; Khodadoust, M.S.; Brown, R.A. Cutaneous T-cell lymphomas with pathogenic somatic mutations and absence of detectable clonal T-cell receptor gene rearrangement: Two case reports. Diagn. Pathol. 2020, 15, 122. [Google Scholar] [CrossRef] [PubMed]
- Cho, W.C.; Gill, P.; Nagarajan, P.; Aung, P.P.; Torres-Cabala, C.A.; Curry, J.L.; Ivan, D.; Lester, L.; Prieto, V.G. Cutaneous Lymphoid Hyperplasia With T-Cell Clonality and Monotypic Plasma Cells Secondary to a Tick Bite: A Hidden Critter and the Power of Deeper Levels. Am. J. Dermatopathol. 2022, 44, 226–229. [Google Scholar] [CrossRef] [PubMed]
- Reeder, M.J.; Wood, G.S. Drug-induced pseudo-Sezary syndrome: A case report and literature review. Am. J. Dermatopathol. 2015, 37, 83–86. [Google Scholar] [CrossRef] [PubMed]
- Miyashiro, D.; Sanches, J.A. Erythroderma: A prospective study of 309 patients followed for 12 years in a tertiary center. Sci. Rep. 2020, 10, 9774. [Google Scholar] [CrossRef] [PubMed]
- Hartmann, S.; Melle, F.; Motta, G.; Agostinelli, C.; Sabattini, E.; Pileri, S.; Hansmann, M.L. Clonal T-cell proliferations occasionally occur in Kikuchi-Fujimoto disease. Hum. Pathol. 2023, 138, 103–111. [Google Scholar] [CrossRef] [PubMed]
- Perry, A.M.; Choi, S.M. Kikuchi-Fujimoto Disease: A Review. Arch. Pathol. Lab. Med. 2018, 142, 1341–1346. [Google Scholar] [CrossRef] [PubMed]
- Yu, F.; Ba, X.; Yang, H.; Huang, K.; Zhang, Y.; Zhang, H.; Xu, L.; Wang, J.; Wang, L.; Wang, Z.; et al. Kikuchi disease with an exuberant proliferation of large T-cells: A study of 25 cases that can mimic T-Cell lymphoma. Histopathology 2023, 82, 340–353. [Google Scholar] [CrossRef] [PubMed]
- Ferrara, J.L.; Levine, J.E.; Reddy, P.; Holler, E. Graft-versus-host disease. Lancet 2009, 373, 1550–1561. [Google Scholar] [CrossRef] [PubMed]
- Simnica, D.; Akyüz, N.; Schliffke, S.; Mohme, M.; VWenserski, L.; Mährle, T.; Fanchi, L.F.; Lamszus, K.; Binder, M. T cell receptor next-generation sequencing reveals cancer-associated repertoire metrics and reconstitution after chemotherapy in patients with hematological and solid tumors. Oncoimmunology 2019, 8, e1644110. [Google Scholar] [CrossRef] [PubMed]
- Simnica, D.; Schliffke, S.; Schultheiß, C.; Bonzanni, N.; Fanchi, L.F.; Akyüz, N.; Gösch, B.; Casar, C.; Thiele, B.; Schlüter, J.; et al. High-Throughput Immunogenetics Reveals a Lack of Physiological T Cell Clusters in Patients With Autoimmune Cytopenias. Front. Immunol. 2019, 10, 1897. [Google Scholar] [CrossRef] [PubMed]
- Malard, F.; Holler, E.; Sandmaier, B.M.; Huang, H.; Mohty, M. Acute graft-versus-host disease. Nat. Rev. Dis. Primers 2023, 9, 27. [Google Scholar] [CrossRef] [PubMed]
- Chang, L.W.; Doan, L.T.; Fields, P.; Vignali, M.; Akilov, O.E. The Utility of T-Cell Clonality in Differential Diagnostics of Acute Graft-versus-Host Disease from Drug Hypersensitivity Reaction. J. Investig. Dermatol. 2020, 140, 1282–1285. [Google Scholar] [CrossRef] [PubMed]
- Winstead, M.; Hill, M.; Amin, Z.; Lugt, M.V.; Chen, X.; Szabolcs, P. Graft-versus-host disease is associated with skewed γδ T-cell clonality after umbilical cord blood transplantation in children with nonmalignant diseases. Cytotherapy 2023, 25, 1091–1100. [Google Scholar] [CrossRef] [PubMed]
- Gong, Q.; Wang, C.; Zhang, W.; Iqbal, J.; Hu, Y.; Greiner, T.C.; Cornish, A.; Kim, J.H.; Rabadan, R.; Abate, F.; et al. Assessment of T-cell receptor repertoire and clonal expansion in peripheral T-cell lymphoma using RNA-seq data. Sci. Rep. 2017, 7, 11301. [Google Scholar] [CrossRef] [PubMed]
- Oon, M.L.; Lim, J.Q.; Lee, B.; Leong, S.M.; Soon, G.S.; Wong, Z.W.; Lim, E.H.; Li, Z.; Yeoh, A.E.J.; Chen, S.; et al. T-Cell Lymphoma Clonality by Copy Number Variation Analysis of T-Cell Receptor Genes. Cancers 2021, 13, 340. [Google Scholar] [CrossRef] [PubMed]
- Yao, W.Q.; Wu, F.; Zhang, W.; Chuang, S.S.; Thompson, J.S.; Chen, Z.; Zhang, S.W.; Clipson, A.; Wang, M.; Liu, H.; et al. Angioimmunoblastic T-cell lymphoma contains multiple clonal T-cell populations derived from a common TET2 mutant progenitor cell. J. Pathol. 2020, 250, 346–357. [Google Scholar] [CrossRef] [PubMed]
- Hamrouni, A.; Fogh, H.; Zak, Z.; Ødum, N.; Gniadecki, R. Clonotypic Diversity of the T-cell Receptor Corroborates the Immature Precursor Origin of Cutaneous T-cell Lymphoma. Clin. Cancer Res. 2019, 25, 3104–3114. [Google Scholar] [CrossRef] [PubMed]
- Iyer, A.; Hennessey, D.; O’Keefe, S.; Patterson, J.; Wang, W.; Salopek, T.; Wong, G.K.; Gniadecki, R. Clonotypic heterogeneity in cutaneous T-cell lymphoma (mycosis fungoides) revealed by comprehensive whole-exome sequencing. Blood Adv. 2019, 3, 1175–1184. [Google Scholar] [CrossRef] [PubMed]
- Harro, C.M.; Sprenger, K.B.; Chaurio, R.A.; Powers, J.J.; Innamarato, P.; Anadon, C.M.; Zhang, Y.; Biswas, S.; Mandal, G.; Mine, J.A.; et al. Sézary syndrome originates from heavily mutated hematopoietic progenitors. Blood Adv. 2023, 7, 5586–5602. [Google Scholar] [CrossRef] [PubMed]
- Iyer, A.; Hennessey, D.; Gniadecki, R. Clonotype pattern in T-cell lymphomas map the cell of origin to immature lymphoid precursors. Blood Adv. 2022, 6, 2334–2345. [Google Scholar] [CrossRef] [PubMed]
- Cieslak, C.; Hain, C.; Rückert-Reed, C.; Busche, T.; Klages, L.J.; Schaper-Gerhardt, K.; Gutzmer, R.; Kalinowski, J.; Stadler, R. Nanopore Sequencing for T-Cell Receptor Rearrangement Analysis in Cutaneous T-Cell Lymphoma. Cancers 2024, 16, 3700. [Google Scholar] [CrossRef] [PubMed]
- Sherwood, A.M.; Desmarais, C.; Livingston, R.J.; Andriesen, J.; Haussler, M.; Carlson, C.S.; Robins, H. Deep sequencing of the human TCRgamma and TCRbeta repertoires suggests that TCRbeta rearranges after alphabeta and gammadelta T cell commitment. Sci. Transl. Med. 2011, 3, 90ra61. [Google Scholar] [CrossRef] [PubMed]
- Kazakov, D.V.; Kutzner, H.; Palmedo, G.; Boudova, L.; Michaelis, S.; Michal, M.; Vanecek, T.; Magro, C.M.; Mukensnabl, P.; Dummer, R.; et al. Primary cutaneous lymphoproliferative disorders with dual lineage rearrangement. Am. J. Dermatopathol. 2006, 28, 399–409. [Google Scholar] [CrossRef] [PubMed]
- Takata, K.; Miyata-Takata, T.; Nishikori, A.; Haratake, T.; Sato, Y. T-cell receptor gamma gene rearrangement analysis of classic Hodgkin lymphoma using a BIOMED-2 assay: A paraffin-embedded tissue analysis of one hundred cases. J. Clin. Exp. Hematop. 2024, 64, 138–143. [Google Scholar] [CrossRef] [PubMed]
- National Comprehensive Cancer Network (NCCN). NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) NCCN Evidence BlocksTM T-Cell Lymphomas Version 4.2024. 2024. Available online: https://www.nccn.org/professionals/physician_gls/pdf/t-cell_blocks.pdf (accessed on 17 September 2024).
- National Comprehensive Cancer Network (NCCN). NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) NCCN Evidence BlocksTM Hodgkin Lymphoma Version 3.2024. 2024. Available online: https://www.nccn.org/professionals/physician_gls/pdf/hodgkin_blocks.pdf (accessed on 17 September 2024).
- National Comprehensive Cancer Network (NCCN). NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) NCCN Evidence BlocksTM Pediatric Hodgkin Lymphoma Version 1.2024. 2024. Available online: https://www.nccn.org/professionals/physician_gls/pdf/ped_hodgkin_blocks.pdf (accessed on 17 September 2024).
- Cheson, B.D.; Fisher, R.I.; Barrington, S.F.; Cavalli, F.; Schwartz, L.H.; Zucca, E.; Lister, T.A.; Alliance, Australasian Leukaemia and Lymphoma Group; Eastern Cooperative Oncology Group; European Mantle Cell Lymphoma Consortium; et al. Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: The Lugano classification. J. Clin. Oncol. 2014, 32, 3059–3068. [Google Scholar] [CrossRef] [PubMed]
- Syrykh, C.; Gorez, P.; Péricart, S.; Grand, D.; Escudié, F.; Cabarrou, B.; Obéric, L.; Ysebaert, L.; Lamant, L.; Laurent, C.; et al. Molecular diagnosis of T-cell lymphoma: A correlative study of PCR-based T-cell clonality assessment and targeted NGS. Blood Adv. 2021, 5, 4590–4593. [Google Scholar] [CrossRef] [PubMed]
- Horna, P.; Weybright, M.J.; Ferrari, M.; Jungherz, D.; Peng, Y.; Akbar, Z.; Tudor Ilca, F.; Otteson, G.E.; Seheult, J.N.; Ortmann, J.; et al. Dual T-cell constant β chain (TRBC)1 and TRBC2 staining for the identification of T-cell neoplasms by flow cytometry. Blood Cancer J. 2024, 14, 34. [Google Scholar] [CrossRef] [PubMed]
- Shi, M.; Jevremovic, D.; Otteson, G.E.; Timm, M.M.; Olteanu, H.; Horna, P. Single Antibody Detection of T-Cell Receptor αβ Clonality by Flow Cytometry Rapidly Identifies Mature T-Cell Neoplasms and Monotypic Small CD8-Positive Subsets of Uncertain Significance. Cytometry B Clin. Cytom. 2020, 98, 99–107. [Google Scholar] [CrossRef] [PubMed]
- Blomme, S.; Nollet, F.; Boeckx, N.; Cauwelier, B.; Snauwaert, S.; Emmerechts, J. Diagnostic utility of the lymphoid screening tube supplemented with TRBC1 for the assessment of T-cell clonality. Int. J. Lab. Hematol. 2023, 45, 496–505. [Google Scholar] [CrossRef] [PubMed]
- Lucero, O.M.; Lee, J.A.; Bowman, J.; Johnson, K.; Sapparapu, G.; Thomas, J.K.; Fan, G.; Chang, B.H.; Thiel-Klare, K.; Eide, C.A.; et al. Patient-Specific Targeting of the T-Cell Receptor Variable Region as a Therapeutic Strategy in Clonal T-Cell Diseases. Clin. Cancer Res. 2023, 29, 4230–4241. [Google Scholar] [CrossRef] [PubMed]
- Varma, G.; Diefenbach, C.S. On the Precipice of a “Rituximab-Like” Era for T-Cell Lymphomas? Clin. Cancer Res. 2023, 29, 4024–4026. [Google Scholar] [CrossRef] [PubMed]
- Ferrari, M.; Righi, M.; Baldan, V.; Wawrzyniecka, P.; Bulek, A.; Kinna, A.; Ma, B.; Bughda, R.; Akbar, Z.; Srivastava, S.; et al. Structure-guided engineering of immunotherapies targeting TRBC1 and TRBC2 in T cell malignancies. Nat. Commun. 2024, 15, 1583. [Google Scholar] [CrossRef] [PubMed]
- Nichakawade, T.D.; Ge, J.; Mog, B.J.; Lee, B.S.; Pearlman, A.H.; Hwang, M.S.; DiNapoli, S.R.; Wyhs, N.; Marcou, N.; Glavaris, S.; et al. TRBC1-targeting antibody-drug conjugates for the treatment of T cell cancers. Nature 2024, 628, 416–423. [Google Scholar] [CrossRef] [PubMed]
Diagnoses of Studied T Cell Neoplasms | Study Purpose | Patients Studied | Clinical Samples Studied | NGS Assays | FL-PCR and FCI Assays, if Used | Relevant Results | References; Country of Study |
---|---|---|---|---|---|---|---|
Advanced stage mycosis fungoides with Sézary syndrome a | To detect MRD by NGS after non-myeloablative allogeneic HSCT in patients with MF and SS | n = 10 (ages >18 <75 y) enrolled in a prospective clinical trial ClinicalTrials.gov #NCT00896493 | PB and skin samples collected before and after HSCT as part of the clinical trial; DNA extracted from PB mononuclear cells | TRB V(D)J NGS [57]; Adaptive Biotechnologies, Seattle, USA | FCI for identifying Sézary cells b | TRB NGS detected clonal CDR3 sequences in all 10 (100%) pre-HSCT {PB (n = 8) and skin (n = 2)}, c including in 6 (60%) patients with no Sézary cells identified by FCI (0.03% to 0.58% of all TRB CDR3 sequences); the percentages of neoplastic clones decreased in all patients immediately post-HSCT | Weng et al., 2013 [58]; USA |
Mycosis fungoides | FL-PCR vs. NGS for clonality evaluation | n = 34 14 males, 20 females; ages 37–76 y | Archived DNA samples from skin biopsies with histologic features of MF; no non-neoplastic cases studied | TRG NGS laboratory-developed test | TRG FL-PCR performed previously with 15 clonal and 19 polyclonal/oligoclonal cases by FL-PCR | 29 (85%) clonal by NGS but only 15 (55%) clonal by FL-PCR; 2 (6%) clonal by FL-PCR but nonclonal by NGS (2.4% and 2.5% sequences by NGS); clonal peaks by FL-PCR if 2-fold signal intensity above Gaussian background | Sufficool et al., 2015 [59]; USA |
CTCL (as defined by [60]), benign inflammatory skin diseases, and healthy donors | To detect T cell clones in early CTCL and distinguish from non-CTCL diseases | n = 110; 104 patients with CTCL (n = 46), and non-CTCL (n = 58) d and 6 healthy donors | DNA from punch biopsies of skin: lesional from CTCL and non-CTCL and from healthy donors | TRB V(D)J NGS and TRG NGS [61]; Adaptive Biotechnologies, Seattle, USA | TRG FL-PCR | NGS showed clones in all 46 (100%) CTCL and had greater sensitivity and specificify than TRG PCR, which was clonal in 27(70)%/39; TRB and TRG NGS were mostly concordant; e a single case of γδ T cell CTCL with a clear clone by TRG NGS was nonclonal by TRB NGS; the most frequent clones by TRB and TRG NGS expressed as a fraction of all nucleated cells in skin f successfully distinguished CTCL from benign non-CTCL and healthy skin | Kirsch et al., 2015 [62]; USA |
T cell prolymphocytic leuekmia (T-PLL) | To detect MRD by NGS as an alternative to detecting by RQ-PCR after allogeneic HSCT | n = 10; 8 males; 2 females; median age at transplant 59 (range 43–72) y | 104 samples: PB (n = 91), BM (n = 10)} and donor PB (n = 3) | TRB V(D)J NGS; EuroClonality–NGS amplicon-based methods | Clone-specific real-time quantitative (RQ) PCR of clonal TRB, TRG, or both rearrangements [63] | 3 patients with the longest follow-up were examined by NGS. In all 3, the diagnostic sample showed 1 or 2 leukemic clonotypes by NGS; MRD evaluation of the leukemic clonotypes by NGS closely followed the kinetics of RQ-PCR-quantified MRD | Sellner et al., 2017 [64]; Germany |
CTCL (primarily MF/SS) | To study CTCL with emphasis on early stage CTCL by NGS | N = 309; included 210 (68%) early-stage CTCL, 54 (17%) stage IIB-III MF, 40 (13%) SS and pre-SS | 309 lesional skin samples | TRB V(D)J NGS; Adaptive Biotechnologies, Seattle, USA | None | The following Vβ families were most frequently used: V20 (13%), V07 (12%), V21 (11%), V06 (9%), V05 (9%), V03 (9%), V10 (5%), V28 (4%), V19 (4%) | de Masson et al., 2018 [65]; USA |
Benign with no history of any LPD; mature T cell neoplasms (MF/SS, cutaneous CD30+ LPD; T-LGLL, ALK- ALCL, PTCL-NOS); mature B cell neoplasms and immune-dysregulation LPDs (DLBCL, EBV+ DLBCL, PT-LPD, EBV+ MCU); and atypical LPDs | TRG FL-PCR vs. TRG NGS for clonality evaluation | N = 41; 22 females and 19 males; median age 59 y (range 9–87) with integrated clinicopathologic diagnoses; 1 MF with SS, 1 MF, 1 cutaneous CD30+ LPD; 2 T-LGLL, 3 ALK- ALCL, 1 PTCL-NOS); mature B cell neoplasms and immune-dysregulation LPDs (3 DLBCL, 1 EBV+ DLBCL, 2 post-transplant LPD, 1 EBV+ MCU); and 4 atypical LPDs | 41 samples (15 PB, 8 BM aspirates, 18 FFPE tissues): 21 benign with no LPD {10 PB, 6 BM, rule out clone: cytopenias (n = 12), other (n = 4), 5 benign lymphoid FFPE tissues}; 9 T cell neoplasms; 7 B cell neoplasms and immune dysregulation-associated LPDs; 4 atypical LPDs (3 EBV+, one suspicious for primary cutaneous CD4+ small/medium T- cell LPD) | TRG NGS; LymphotrackTM, Invivoscribe Inc., San Diego, USA | Two TRG FL-PCR assays (1-tube [66] g and 2-tube) (Invivoscibe Inc., San Diego, USA) | TRG NGS provided an accurate assessment of all polyclonal, oligoclonal, and clonal T cell populations in 100% of cases diagnosed by integrated clincopathologic features. False positive FL-PCR: 3 (30%)/10 benign PB; 1 (16.6%)/6 benign BM aspirate cases; 1 (25%)/4 atypical LPD cases. False negative FL-PCR: 3 (30%)/10 benign PB cases; 3 (60%)/5 PB cases in patients with LPDs (1 MF, 1 PT-LPD, 1 EBV+ MCU); 1 (25%) (1 ALK-negative ALCL) in 4 mature T cell lymphomas; 1 PT-LPD in FFPE tissue (~3.5% clone by NGS); and 1 primary cutaneous CD4+ small/medium T cell LPD (25%) of 4 atypical LPDs | Kansal et al., 2018 [19]; USA |
Clinical concern for a diagnosis of CTCL; cases in four histologic diagnoses | FL-PCR vs. NGS for clonality evaluation | N = 100 (N = 25 in each of the four histologic categories: ‘definitive CTCL’, ‘atypical lymphoid infiltrate, concerning for CTCL’, ‘atypical lymphoid infiltrate, favor reactive’ and ‘reactive lymphoid infiltrate’) | 100 skin biopsies and concurrent PB samples examined by FL-PCR and NGS; most PB samples also analyzed by FCI | TRB V(D)J NGS; ImmunoSEQ, Adaptive Biotechnologies, Seattle, USA | TRG FL-PCR; FCI for PB samples | In skin, 100% diagnostic specificity of NGS vs. 88% by TRG PCR, with similar diagnostic sensitivity (68% vs. 72% by PCR) and accuracy (84% vs. 80%). In PB samples, TRG FL-PCR and FCI were the least useful for diagnosis. NGS showed non-identical sequences in some identically sized peaks by TRG FL-PCR in concurrent skin and PB samples. | Rea et al., 2018 [67]; USA |
T cell prolymphocytic leukemia (T-PLL) | Compare TRB NGS (BIOMED-2 Vβ-Jβ primers) with Vβ FCI | N = 80; 47 (59%) males, 33 (41%) females; median age 64.5 y (range 38–84); | 80 diagnostic PB samples | TRB V(D)J NGS using BIOMED-2 Vβ-Jβ primers | Vβ expression by FCI [68,69] | A dominant Vβ domain usage was detected by FC1 in only 41 (51%)/80 samples, but clonality was suspected in all samples by FCI. In 12 (15%) cases, NGS identified the clone missed by FCI. Overall, NGS and FCI results were concordant in 61 (76%) of 80 samples. | Kotrova et al., 2018 [70]; Germany, Czech Repubic |
CTCL | Quantitate tumor burden in matched skin and PB samples from patients with CTCL and reactive conditions | N = 46; 23 (16 males and 7 females) with histological and clinical diagnosis of CTCL and 23 with reactive conditions | Skin biopsy and concurrent PB samples assessed by TRB NGS in 46 patients | TRB V(D)J NGS; ImmunoSEQ, Adaptive Biotechologies, Seattle, WA | None | 7 (30%) CTCL showed identical top frequency skin and PB (1.8–86.8%) clones; 16 (70%) CTCL showed non-identical skin (0–79.4%) and PB clones. In 16 (70%)/23 reactive conditions, NGS detected the top frequency skin clone in PB (0.0012–0.71%); none of these 23 patients had a larger skin clone in PB. | Wang et al., 2019 [71]; USA |
Reactive T cell lymphocytosis or lymphoid hyperplasia, T cell NHL-NOS, ALCL, AITL, PTCL-NOS, T cell PLL, T-LGLL, MF, SS, T-ALL/T-LBL, B-NHL and B-ALL, hypereosinophilic syndrome, HL, MDS, non-hematological diseases, and GVHD | FL-PCR vs. NGS for clonality evaluation | N = 121; {Reactive T cell lymphocytosis or lymphoid hyperplasia (n = 37), T cell NHL-NOS (n = 3), ALCL (n = 2), AITL (n = 1), PTCL-NOS (n = 2), T-PLL (n = 1), T-LGLL (n = 5), MF (n = 1), SS (n = 1), T-ALL/T-LBL (n = 9), B-NHL (n = 6), B-ALL (n = 2), hypereosinophilic syndrome (n = 7), HL (n = 1), MDS (n = 2), non-hematological diseases (n = 4), GVHD (n = 1), unavailable final diagnoses (n = 36)} | 121 diagnostic samples suspicious for a T cell LPD {85 fresh samples (11 PB, 33 BM, 37 lymph nodes, 4 skin biopsies) with definitive diagnoses established by clinical, morphological, and immunophenotypic data, and 36 referred cases of FFPE tissue sections with unavailable final clinical diagnosis)} | TRG NGS; LymphotrackTM Dx | TRG FL-PCR | In 94.4% of FFPE, NGS showed reliable results despite unavailable final diagnosis; 55.6% (n = 20/36) of FFPE cases non-interpretable by FL-PCR. False positive FL-PCR cases: (1) 1 normal case monoclonal by PCR confirmed as polyclonal by NGS; (2) in 14 fresh samples with FL-PCR monoclonal in a polyclonal background, NGS clarified a polyclonal pattern (n = 12; normal, B-NHL, and benign), polyclonal with minor clones of uncertain significance (n = 1), or monoclonal with only 9.6% clonal sequences (n = 1, B-NHL). False negative FL-PCR: in 2 of 42 cases polyclonal by FL-PCR (1 benign; one B cell lymphoma), NGS showed clonal sequences (9.2% and 6.3% of total). | Nollet et al., 2019 [51]; Belgium |
Advanced MF and SS after allogeneic HSCT | To monitor MRD after HSCT by NGS | N = 35, 13 MF, 22 SS; 21 males, 14 females; median age 60 (20–74) y in a clinical trial (#NCT00896493) | Diagnostic samples available for NGS (in n = 30); concurrent PB and skin samples evaluated for MRD | TRB V(D)J NGS; Adaptive Biotechnologies, Seattle, USA | None | In PB samples, NGS identified molecular remission (MR), defined as undetectable malignant clone with a sensitivity of 1 in 200,000 nuclear cells. | Weng et al., 2020 [72]; USA |
T cell leukemia/lymphoma, hematologic malignancy (e.g., AML, MDS), non-malignant inflammatory disorder | FL-PCR vs. NGS for clonality evaluation | N patients unavailable; 72 specimens, majority stated to be from the CTCL clinic | 72 samples (20 BM, 21 PB, 14 fresh tissues; 17 FFPE tissues) | TRG NGS; LymphotrackTM | TRG FL-PCR | 28 (77.8%) of 36 cases with a T cell neoplasm were clonal by NGS; 3 specimens with false negative PCR (having clinicopathologic diagnoses of T cell leukemia/lymphoma) were clonal by NGS. NGS showed a trackable sequence in 11 patients with ≥2 specimens evaluated for TRG clonality. | Lay et al., 2020 [50]; USA |
Relapsed or refractory MF/SS treated with mogamulizumab (anti-chemokine receptor type 4 antibody) [73] | Study skin biopsies of mogamulizumab-associated rash examined by NGS or FL-PCR | 19 patients who developed mogamulizumab-associated rash; excluded biopsies with dominant TRG or TRB sequences | 52 biopsies from 19 patients who developed mogamulizumab-associated rash | TRB V(D)J NGS and TRG NGS; Adaptive Biotechnologies, Seattle, USA [58,62] | TRB and TRG FL-PCR [74] used in only 6 specimens where NGS was not used | 20 of 46 biopsies analyzed by NGS showed low levels of neoplastic T cell receptor sequences identified before treatment; in 38 of 43 biopsies, intraepidermal lymphocytes showed an inverted CD4:CD8 ratio ≤1:1 by immunohistochemistry. | Wang et al., 2020 Dec [75]; USA |
CTCL (primarily early or partially treated disease) and various non-CTCL conditions h | FL-PCR vs. FCI vs. NGS for T cell clonality | N = 55 from the CTCL clinic; 34 males, 21 females; ages 21–85 years; integrated diagnosis of CTCL confirmed in N = 35 after clinical, histologic, immunophenotypic and T cell clonality evaluation | In 102 skin biopsies: 59 FCI tests in n = 53 patients, 50 FL-PCR tests in n = 50, 26 NGS tests in n = 23; In 51 PB samples for FCI (in n = 51), 50 FL-PCR (in n = 50), 16 NGS tests (in n = 16); 15 concurrent FL-PCR and NGS | TRB V(D)J NGS; Adaptive Biotechnologies, Seattle, USA | T cell receptor BIOMED-2 FL-PCR using targets Vβ, Dβ, Jβ, Vγ, and Jγ (TRB and TRG) | FL-PCR overall sensitivity 61% in skin and 40% in PB; FL-PCR overall specificity 64% in skin and 61% in PB; TRB NGS overall sensitivity similar (60%) but greater (87%) overall specificity; of note, 28% and 21% of skin samples were insufficient for FL-PCR and FCI, respectively. i Specificity in PB: 100% for TRB NGS, 70% for FCI, 62.5% for FL-PCR. j | Gibbs et al., 2021 [76]; USA |
Various: MF, AITL, PTCL, T-LGLL, other TCL, HL, MZL, CLL, other BCL, reactive, cytopenia, eosinophilia, leukocytosis, other | FL-PCR vs. NGS for clonality evaluation | N patients unavailable {MF/SS (18%), AITL (7%), PTCL (7%), T-LGLL (7%), other TCL (7%), HL (5%), MZL (2%), CLL (1%), other BCL (2%), reactive (5%), cytopenia (21%), eosinophilia (4%), leukocytosis (2%), other (1%), unknown (11%)} | 101 DNA samples (48 PB, 46 FFPE tissues, 7 BM aspirates) previously analyzed by TRG FL-PCR as part of the diagnostic workup for suspicion of T cell malignancy or LPD | TRG NGS; LymphotrackTM | TRG FL-PCR (2-tube BIOMED-2 PCR assay lacking the JγP primer k) | TRG NGS: accuracy 83%; analytical specificity 100%; discordant results between FL-PCR and NGS reported in 25 samples {17 (68%)/25 PB, 1 BM aspirate, 7 FFPE tissues}: 17 samples (11 PB, 6 FFPE) clonal by FL-PCR with discordant results by NGS {9 false positive PCR (8 PB, 1 FFPE), 5 false negative NGS l (2 T-LGLL, 1 PTCL, 2 AITL), 2 false oligoclonal NGS (1 PTCL, 1 AITL), 1 diagnosis unknown}; 8 samples (6 PB, 1 BM, 1 FFPE) oligoclonal by FL-PCR were polyclonal by NGS. m | Ho et al., 2021 [49]; USA |
T cell and B cell malignancies and reactive lesions | NGS DNA Capture assay vs. standard testing for B cell and T cell malignancy cases submitted by ten EuroClonality–NGS laboratories | N patients unavailable; 204 B- and 76 T cell malignancies {42 T-ALL, 1 T-LBL, and 33 mature T neoplasms (13 ALCL, 9 AITL, 3 MF, 3 SS, 2 EATL, 1 C-ALCL, 1 PTCL-NOS, 1 intestinal TCL-NOS)}; 21 non-neoplastic | DNA from 76 T cell malignancies {42 T-ALL, 1 T-LBL, and 33 mature T neoplasms; 9 HMW DNA, 67 FFPE}; 21 reactive lesions; 14 LPD cell lines; 4 cell line blends | EuroClonality–NGS DNA Capture assay to detect T cell clonality at TRA, TRB V(D)J and TRB DJ, TRD, or TRG loci | The original BIOMED-2 T cell receptor FL-PCR results from the ten participating laboratories for all cases | The NGS assay detected TCR clonality, i.e., ≥1 clonal rearrangements at TRA, TRB, TRD, or TRG loci, in 71 (97%) of 73 T cell malignancies (1 T-ALL and 1 ALCL negative for a clone by FL-PCR and NGS). All 7 mature TCLs clonal by TRG NGS but only 1 clonal by TRG FL-PCR; 6 of 7 clonal by TRB NGS, but TRB FL-PCR clonal in only 3 (43%) of 7. | Stewart et al., 2021 [77]; Europe |
CTCL and non-neoplastic | To evaluate the role of TRB NGS in diagnosing CTCL; NGS data from 2013 to 2020 analyzed retrospectively | 144 patients: 101 CTCL {52 MF, 35 SS, 8 other TCL, 6 CD30+ LPD (3 ALCL, 3 LyP)} [78] and 43 non-neoplastic | Skin samples from 101 CTCL and 43 non-neoplastic cases; also PB in 9 cases of SS | TRB V(D)J NGS; Adaptive Biotechnologies, Seattle, USA | TRG FL-PCR analyzed PB and skin in 9 cases of SS | TRB NGS diagnostic specificity for CTCL: 95% and 100%; sensitivity 89% and 50%, with the top T cell clones comprising 5% and 25%, respectively, of all T cells in skin. PB in 1 (11%) SS case was non-clonal by FL-PCR; in that case, skin and PB were clonal by NGS. | Zimmerman et al., 2021 [79]; France |
Refractory or relapsed MF or SS treated with mogamulizumab | To describe the diagnosis and treatment of mogamulizumab-associated skin rash in patients with CTCL | N = 24; including 13 males, 11 females; median age 65.1 (30.4–88.9) years; 17 of 24 developed a rash after treatment with mogamulizumab | Skin biopsies in 14 of those 17 patients who developed a rash after treatment; skin and PB examined by FL-PCR and NGS | Laboratory-developed TRG and TRB V(D)J NGS assays in skin and PB (performed after [19,51] were published) | Laboratory-developed TRG and TRB FL-PCR in skin and PB (completed before [19,51] were published) | All 17 patients showed TRG or TRB clones in skin, PB, or lymph node specimens before treatment by FL-PCR or NGS. After treatment with mogamulizumab, 12 (85.7%)/14 skin and 9 (75%)/12 PB samples examined were absent for a T cell clone. | Trum et al., 2022 [80]; USA |
Clinical and histopathologic diagnoses of pityriasis lichenoides | To evaluate NGS in pediatric patients with pityriasis lichenoides | 12 pediatric patients (5 females, 7 males; ages 3–16 years) | 18 biopsy specimens from 12 patients; five of the 12 patients had two concurrent skin biopsies from different anatomic sites | TRG NGS and TRB V(D)J NGS; LymphoTrackTM | None | 7 (58%)/12 patients showed T cell clones: TRG in n = 6 (50%); TRB in n = 3 (25%), including n = 2 TRG clonal}; in n = 5 patients with concurrent biopsies, matching TRB clones in n = 2, no TRG or TRB clone in either biopsy in n = 2, and only TRG clonal in 1 of 2 biopsies in n = 1; discordant TRG and TRB results in n = 5 (42%)/12 patients (TRG clonal and TRB nonclonal in n = 4; TRB clonal but TRG nonclonal in n = 1) | Raghavan et al., 2022 [81]; USA |
Folliculotropic variant of MF, presenting with the plaque stage | Evaluation of tumor clone frequency by TRB NGS | 41 patients, including 24 with progressive disease, including 16 dead of the disease | 46 residual FFPE or frozen skin biopsies at diagnosis from 41 patients | TRB V(D)J NGS; Adaptive Biotechnologies, Seattle, USA | None | NGS in 41 patients showed a tumor clone frequency of 0.95% to 72.01%, with a median of 11.00% | Van Santen et al., 2022 [82]; The Netherlands |
Early-stage MF | To study TRG NGS patterns in successive and synchronous skin lesions | 7 patients wth early-stage MF | 17 skin biopsies in 7 patients (7 sequential in 3 patients, 11 synchronous in 5 and both in 1 patient) | TRG NGS; BIOMED-2- based laboratory-developed assay | Previously assessed by BIOMED-2 TRG and TRB FL-PCR | NGS identified identical dominant TRG rearrangements in all samples spatially and temporally in a given patient, but NGS was minimally more sensitive than FL-PCR. | Bozon et al., 2022 [83]; France |
T cell malignancies | LymphotrackTM MRD assays compared with ClonoSEQ MRD assays for routine MRD monitoring | 60 patients {37 CTCL (MF, SS), 8 T-ALL, 6 B-ALL, 2 PTCL, 2 AITL, 1 adult T cell lymphoma, 2 T-LGLL, 1 T cell lymphoma NOS, and 1 T-PLL} | 69 follow-up samples (65 PB, 4 BM) analyzed from 60 patients; initial neoplastic clone identification by ClonoSEQ | NGS TRB V(D)J, and TRG MRD; LymphotrackTM compared with ClonoSEQ TRG and TRB V(D)J MRD (Adaptive Technologies, Seattle, USA) | Concurrent FCI in 32 (46%) samples; no FL-PCR assays | Both manufacturers’ TRG MRD assays and FCI performed in 31 samples: 19 (31%) MRD+ by all 3 assays, and 12 (38.7%) MRD+ by both TRG NGS assays but negative by FCI. Both TRB MRD assays and FCI performed in 28 samples: 17 (60.7%) MRD+ by all 3 assays, 11 (39%) MRD+ by both TRB NGS asays but negative by FCI. | Tung et al., 2023 [84]; USA |
Mycosis fungoides | To study neoplastic intraclonal CDR3 variants in MF by NGS | N = 3 (2 males and one female) described from a cohort of 21 patients (of 60 reviewed) | 23 (22.5%) of 102 MF skin biopsies in 21 patients with ≥2 dominant T cell clones | TRB V(D)J NGS; Adaptive Biotechnologies, Seattle, USA | None | 6 (26%) of these 23 biopsies from 3 patients showed single base substitutions in the TRB sequences representing intraclonal variations. | Gleason et al., 2023 [85]; USA |
MF without PB involvement, defined according to Olsen et al., 2022 [86] | To study T cell clones in skin and PB samples in patients with MF limited to skin | N = 60 with confirmed histopathological diagnosis of MF and no PB involvement; 50 (83%) of 60 early-stage (IA-IIA) MF | PB and lesional skin biopsies in 60 patients | TRB V(D)J NGS; Adaptive Biotechnologies, Seattle, USA | None | 17 (28%) of 60 patients had a dominant T cell clone in PB by NGS; the clone was identical in skin and PB in 3 (18%) and discordant between skin and PB in 14 (82%, early-stage MF). | Joffe et al., 2023 [87]; USA |
Classic Hodgkin’s lymphoma at diagnosis and relapse | To study the clonal relationship at diagnosis and relapse | N = 60; median age 26 (4–76) y {ages ≤ 18 y (n = 18); >18 y (n = 42)}; 37 males, 23 females | 130 tissue samples (99 FFPE, 31 fresh frozen); paired tissue samples at diagnosis and relapse in N = 60; T cell clonality in 125 samples in 57 patients | EuroClonality–NGS amplicon-based assays for IGH, IGK, TRB V(D)J, TRB DJ, TRG [39,40,42] | None | 14 (11%)/125 samples in 12 (21%)/57 patients showed T cell clones at diagnosis (n = 5) and relapse (n = 9); 7 (50%)/14 samples in 5 patients were retrospectively diagnosed as TCL based on retrospective pathology review and TCL-associated somatic mutations. n | Van Bladel et al. 2023 [46]; Europe |
Mycosis fungoides with treatment-related skin rash | Treatment-related skin rash vs. disease progression | N = 3; two females ages 88 y and 90 y, and one male, age 44 y | PB and skin biopsies {mogamulizumab-associated rash (n = 2); mechlorethamine gel dermatitis (n = 1) | TRB V(D)J NGS; Adaptive Biotechnologies, Seattle, USA | FCI in PB samples (n = 2) | A T cell clone similar to the neoplastic clone at diagnosis was absent by NGS in skin and PB of all three patients; no PB involvement by FCI (n = 2) | Bhatti et al., 2023 [88]; USA |
Mature T cell neoplasms, all αβ subtype, CD3+: SS, AITL, ALK-negative ALCL, MF, nodal PTCL-TFH, PTCL-NOS, SCPLTCL, T-LGLL, T-PLL, unclassifiable | FCI for T cell receptor β constant chain 1 (TRBC1) compared with TRB NGS | N = 57 (24 females, 34 males; median age 58 y) 11 SS, 1 AITL, 1 ALK-negative ALCL, 3 MF, 3 nodal PTCL, TFH subtype, 6 PTCL-NOS, 2 SCPLTCL, 1 T-LGLL, 2 T-PLL, 1 unclassifiable | 90 samples (48 PB, 37 BM, and 5 lymph nodes), including follow-up samples with variable tumor burden from 57 patients | TRB V(D)J NGS LymphotrackTM assay in 38 samples with available DNA | TRBC1 by FCI performed in 90 samples | FCI for TRBC1 confirmed T cell clones in 37 (97%)/38 samples involved by a mature T cell neoplasm in 17 patients but also identified T cell clones of uncertain significance in 9 (100%)/9 non-neoplastic samples (5 PB, 4 BM) in 6 (10%) patients. In 17 samples negative for disease, FCI showed polyclonal T cells, and TRB NGS detected T cell clonal disease in 6 patients in 6 (35%)/17 samples. o | Nguyen et al., 2024 [89]; Australia |
Anaplastic lymphoma kinase (ALK)+ anaplastic large-cell lymphoma | Use real-time quantitative reverse transcriptase PCR to diagnose and monitor MRD in ALK+ ALCL | 96 patients {age range 2–80 (median 16) years} with ALK+ ALCL | TRB NGS and TRG NGS used in 23 patients in diagnostic FFPE or frozen tissues | TRB V(D)J and TRG EuroClonality–NGS amplicon-based assays [39] p | None | 18 (78%)/23 cases showed at least 1 T cell clone; both TRB and TRG were clonally rearranged in 16 (70%); 1 case each was clonal only for TRG and TRB; no clone by TRG or TRB (in n = 5). All TRG-clonal cases had 2 major clones; 13 (72%)/17 TRB-clonal cases had 1 major clone; 2 cases showed 2 TRB clones. | Kalinova et al., 2024 [90]; The Czech Republic |
Nodal T-follicular helper cell lymphoma, angioimmunoblastic type (or AITL), BM involvement | To study clonal hematopoisis in BM biopsies of patients with AITL | N = 22; {at diagnosis of lymphoma, median age 68 (range 47–83 years); 10 males, 12 females}; 29 BMs in 22 studied | TRB NGS used in 11 BM biopsies from 7 patients with AITL; FFPE tissues from the diagnostic lymph node and clinical staging BM biopsies | OncomineTM TCR beta-short read (SR) NGS assay for TRB V(D)J (Thermofisher Scientific) [91] | None | Neoplastic lymph nodes in all 7 patients and BMs with definite neoplastic infiltration (n = 4) showed high proportions (32–80%) of clonal neoplastic sequences. In 3 patients, 4 BMs negative (n = 3) or suspicious for lymphoma (n = 1) by morphology showed <1.5% of sequences similar to the neoplastic clone by NGS. q | Harland et al., 2024 [92]; Germany |
PB Cases a | TRG FL-PCR 2-Tube | v2.0 TRG FL-PCR Single Tube | TRG NGS b |
---|---|---|---|
PB3 | No clone | No clone | Clone, ~2.4% |
NLPD1 | Clonal | Not diagnostic for clone; few target cells | No clone, few reads |
NLPD3 | Clonal | Not diagnostic for clone | Clone, ~5.5% |
NLPD4 | Borderline clone | No clone | No clone |
NLPD6 | Clonal | Not diagnostic for clone | Clone, ~7% |
NLPD7 | Clonal | Not diagnostic for clone | No clone |
Type of Sample | LPD Diagnosis | TRG PCR 2-Tube | v2.0 PCR Single Tube | TRG NGS a |
---|---|---|---|---|
PB | MF and SS | Clonal | Clonal | Clonal, at least ~12% |
PB | EBV+ MCU | Non-diagnostic | Not diagnostic for clone | Clonal, at least 8% |
PB | MF | Clonal | Not diagnostic for clone | Clonal, at least 3% |
PB | Cutaneous CD30+ LPD | Clonal | Clonal | Clonal, at least 64% |
PB | Post-transplant LPD | Oligoclones | Oligoclones | Clonal, at least 28% |
BM | T-LGLL | Clonal | Clonal | Clonal, at least 17% |
BM | T-LGLL | Clonal | Clonal | Clonal, at least 12% |
FFPE Cases a | Diagnosis | TRG FL-PCR 2-Tube | V2.0 FL-PCR Single Tube | TRG NGS b |
---|---|---|---|---|
Mature T cell lymphomas (n = 4) | ||||
LPD8 | ALK-negative ALCL, recurrence | Non-diagnostic | Clonal, biallelic | Clonal, ~66%, biallelic |
LPD9 | Peripheral T cell lymphoma, NOS | NA | Clonal, biallelic | Clonal, ~60%, monoallelic |
LPD10 | ALK-negative ALCL | NA | Clonal, biallelic | Clonal, ~44%, monoallelic |
LPD11 | ALK-negative ALCL | NA | Clonal, biallelic | Clonal, ~69%, biallelic |
Mature B cell neoplasms and immune dysregulation LPDs (n = 5) | ||||
LPD12 | DLBCL, non-GC B cell phenotype | NA | Clonal | Clone, ~8% |
LPD13 | EBV+ positive DLBCL, NOS | NA | No clone, few target cells | Polyclonal, no definite clone in few target cells |
LPD14 | DLBCL, GC B cell phenotype | NA | Not diagnostic for clone | Polyclonal, no clone, few target cells |
LPD15 | DLBCL, non-GC B cell phenotype | NA | No clone, few target cells | Few target cells, no clone |
LPD16 | Post transplant LPD, plasmacytic type | No T cell clone | One borderline clonal peak | Clone, ~3.5%, ample target cells |
Atypical LPDs (n = 4) | ||||
ALPD1 | Atypical LPD, EBV+ | Clonal | Not diagnostic for clone | Borderline expansion, no definite clone |
ALPD2 | Atypical LPD suspicious for but not diagnostic of primary cutaneous CD4+ small/medium T cell LPD | Clonal | Not diagnostic for clone | Clonal, at least 12% |
ALPD3 | Atypical LPD, EBV+ | No clone | Not diagnostic for clone | Polyclonal, no clone |
ALPD4 | Atypical LPD, post-transplant, EBV negative | No clone | Polyclonal | Polyclonal, no clone |
BIOMED-2 FL-PCR Results a [77] | EuroClonality–NGS DNA Capture Assay Results [77] | ||||||
---|---|---|---|---|---|---|---|
Case # | Clonal for TRG or TRB | Polyclonal for TRG, TRB, or both | TRD | TRG | TRB | TRA | Translocations |
Anaplastic large-cell lymphoma (n = 5 cases) | |||||||
1 | None | Both TRG and TRB polyclonal | Clonal | Clonal | Clonal | Nonclonal | None |
2 | TRB clonal (V-J) | TRG polyclonal | Clonal | Clonal | Clonal | Clonal | ALK |
3 | None | TRG and TRB (V-J and D-J) polyclonal | Clonal | Clonal | Clonal | Nonclonal | ALK |
4 | TRB clonal (D-J) | TRG and TRB V-J polyclonal | Clonal | Clonal | Clonal | Nonclonal | ALK |
5 | TRB clonal (D-J) | TRG and TRB V-J polyclonal | Nonclonal | Clonal | Clonal | Clonal | ALK |
Angioimmunoblastic T cell lymphoma (n = 2 cases) | |||||||
1 | TRG clonal | TRB (V-J and D-J) polyclonal | Nonclonal | Clonal | Clonal | Clonal | None |
2 | None | Both TRG and TRB polyclonal | Nonclonal | Clonal | Nonclonal | Clonal | None |
TRB by EuroClonality NGS: Clonal or Polyclonal | TRG V-J by EuroClonality NGS: Clonal or Polyclonal | Sample Conclusion for T Cell Clonality | |||
---|---|---|---|---|---|
Case a | Sample at Diagnosis or Relapse | TRB V-(D)-J | TRB D-J | ||
1 | Diagnosis | Clonal | Polyclonal | Clonal | Clonal |
Relapse | Clonal a | Polyclonal | Clonal b | Clonal | |
5 | Diagnosis | Polyclonal | Clonal | Clonal c | Clonal |
Relapse | Polyclonal | Clonal | Clonal b | Clonal | |
11 | Diagnosis | Polyclonal | Clonal c | Clonal c | Polyclonal |
First Relapse | Polyclonal | Clonal b | Clonal b | Clonal | |
Second Relapse | Polyclonal | Polyclonal | Polyclonal | Polyclonal | |
17 | Diagnosis | Polyclonal | Polyclonal | Polyclonal | Polyclonal |
First Relapse | Polyclonal | Polyclonal | Polyclonal | Polyclonal | |
Second Relapse | Clonal | Polyclonal | Polyclonal | Clonal | |
23 | Diagnosis | Not evaluable | Polyclonal | Clonal c | Polyclonal |
First Relapse | Clonal | Polyclonal | Clonal | Clonal | |
Second Relapse | Polyclonal | Polyclonal | Polyclonal | Polyclonal | |
26 | Diagnosis | Clonal | Polyclonal | Polyclonal | Clonal |
First Relapse | Polyclonal | Polyclonal | Polyclonal | Polyclonal | |
Second Relapse | No specific product | Polyclonal | Polyclonal | Polyclonal | |
27 | Diagnosis | Polyclonal | Polyclonal | Polyclonal | Polyclonal |
Relapse | Clonal | Clonal | Clonal | Clonal | |
34 | Diagnosis | Polyclonal | Clonal | Clonal | Clonal |
Relapse | Polyclonal | Polyclonal | Polyclonal | Polyclonal | |
39 | Diagnosis | Polyclonal | Polyclonal | Polyclonal | Polyclonal |
Relapse | Polyclonal | Clonal | Polyclonal | Clonal | |
41 | Diagnosis | Polyclonal | Polyclonal | Polyclonal | Polyclonal |
Relapse | Polyclonal | Clonal | Clonal | Clonal | |
50 | Diagnosis | Not performed | |||
First Relapse | Clonal | Polyclonal | Polyclonal | Clonal | |
Second Relapse | Polyclonal | Polyclonal | Polyclonal | Polyclonal | |
60 | Diagnosis | Clonal | Polyclonal | Polyclonal | Clonal |
Relapse | Polyclonal | Polyclonal | Polyclonal | Polyclonal |
TRG and TRB NGS Assays | TRB Targets | Types of Samples, N Cases Compared, and Diagnoses Studied | Comparative TRG and TRB Interpretation Summary | |
---|---|---|---|---|
Kirsch et al., 2015 [62] | Adaptive Biotechnologies, Seattle, USA | V(D)J | Skin; N = 110, in CTCL and non-CTCL diseases, and healthy donors | Both assays similar except in one case; only TRG NGS detected the γδ T cell neoplasm |
Stewart et al., 2021 [77] | EuroClonality–NGS DNA Capture Assay, Europe | V(D)J and DJ | FFPE tissues and high molecular weight DNA, N = 7, T cell lymphomas (5 ALCL, 2 AITL) | TRG superior to TRB in the 7 cases with separate results available for TRB and TRG NGS; see Table 7 |
Trum et al., 2022 [80] | Laboratory-developed NGS assays, USA | V(D)J | Skin and PB; N = 14, relapsed or refractory CTCL or SS treated with mogamulizumab | Assays were not compared; comparing assays was not the study’s purpose |
Raghavan et al., 2022 [81] | LymphotrackTM (Invivoscribe Inc., San Diego, USA) | V(D)J | Skin biopsies, N = 12, pediatric pityriasis lichenoides | NGS is not helpful in this diagnosis (results can be clonal or non-clonal) |
Tung et al., 2023 [84] | Adaptive Biotechnologies, Seattle, USA, and LymphotrackTM (Invivoscribe Inc., San Diego, USA) | V(D)J in both assays | PB and bone marrow aspirate samples; N = 28; various T cell neoplasms examined for minimal/measurable residual disease | TRG NGS superior to TRB NGS; see above text in this section |
Van Bladel et al., 2023 [46] | EuroClonality–NGS amplicon-based assay | V(D)J and DJ | FFPE and fresh frozen tissues, N = 60, CHL at diagnosis and relapse only | TRB NGS better than TRG NGS in 14 samples; a see Table 8 |
Kalinova et al., 2024 [90] | EuroClonality–NGS amplicon-based assay | V(D)J | FFPE and fresh frozen tissues; N = 23; all ALCL, anaplastic lymphoma kinase (ALK) positive only | Both assays similar; TRG and TRB NGS identified one case each as clonal that was not identified as clonal by the other assay |
Authors, Year Published, Country | Patient Age, Sex | Clinical Significance of the Case Reports |
---|---|---|
Hwang et al., 2021 [103]; USA | 55 years, female | The presence of a false positive T cell clone by FL-PCR led to the misdiagnosis of a benign condition as a hepatosplenic γδ T cell lymphoma [103], which is an aggressive lymphoma [104]. NGS for T cell receptor clonality showed the polyclonal nature of the T cells and a hematopoietic stem cell transplant was avoided in the patient [103]. |
Zhang et al., 2016 [105]; UK | 25 years, male | Acute Epstein–Barr virus (EBV) infection can present with histopathologic features and a clonal T cell population by FL-PCR to mimic a peripheral T cell lymphoma. |
Rojansky et al., 2020 [106]; USA | 38 and 69 years, males | Two cases of cutaneous T cell lymphoma wherein T cell receptor NGS and T cell receptor FL-PCR did not reveal a T cell clone, and only testing for somatic gene mutations revealed a clone and the neoplastic nature of these cases. |
Cho et al., 2022 [107]; USA | 81 years, female | Cutaneous lymphoid hyperplasia due to a tick bite with a clonal T cell population by FL-PCR and lambda light chain-restricted plasma cells mimicking primary cutaneous CD4+ small/medium T cell lymphoproliferative disorder and Borrelia-associated primary cutaneous marginal zone B cell lymphoma, respectively; the correct diagnosis was established by deeper histologic tissue sections showing tick parts. |
Reeder and Wood, 2015 [108]; USA | 51 years, male | Erythroderma and pseudo- Sézary syndrome due to antihypertensive medications occurred in the patient with histologic, flow cytometric immunophenotypic, and molecular clonality findings by FL-PCR mimicking neoplastic disease in skin and peripheral blood; the atypical findings resolved after stopping the medications, emphasizing the importance of clinical history in accurate diagnosis. |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the author. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kansal, R. Is It Time to Assess T Cell Clonality by Next-Generation Sequencing in Mature T Cell Lymphoid Neoplasms? A Scoping Review. J. Mol. Pathol. 2025, 6, 2. https://doi.org/10.3390/jmp6010002
Kansal R. Is It Time to Assess T Cell Clonality by Next-Generation Sequencing in Mature T Cell Lymphoid Neoplasms? A Scoping Review. Journal of Molecular Pathology. 2025; 6(1):2. https://doi.org/10.3390/jmp6010002
Chicago/Turabian StyleKansal, Rina. 2025. "Is It Time to Assess T Cell Clonality by Next-Generation Sequencing in Mature T Cell Lymphoid Neoplasms? A Scoping Review" Journal of Molecular Pathology 6, no. 1: 2. https://doi.org/10.3390/jmp6010002
APA StyleKansal, R. (2025). Is It Time to Assess T Cell Clonality by Next-Generation Sequencing in Mature T Cell Lymphoid Neoplasms? A Scoping Review. Journal of Molecular Pathology, 6(1), 2. https://doi.org/10.3390/jmp6010002