Early detection of T-cell lymphoma with T follicular helper by RHOA mutation analysis by Rachel Dobson, Peter Y. Du, Lívia Rásó-Barnett, Wen-Qing Yao, Zi Chen, Calogero Casa, Hesham EI-Daly, Lorant Farkas, Elizabeth Soilleux, Penny Wright, John W. Grant, Manuel Rodriguez-Justo, George A. Follows, Hala Rashed, Margarete Fabre, E. Joanna Baxter, George Vassiliou , Andrew Wotherspoon, Ayoma D. Attygalle, Hongxiang Liu, and Ming-Qing Du

Haematologica 2021 [Epub ahead of print]

Citation: Rachel Dobson, Peter Y. Du, Lívia Rásó-Barnett, Wen-Qing Yao, Zi Chen, Calogero Casa, Hesham EI-Daly, Lorant Farkas, Elizabeth Soilleux, Penny Wright, John W. Grant, Manuel Rodriguez-Justo, George A. Follows, Hala Rashed, Margarete Fabre, E. Joanna Baxter, George Vassiliou, Andrew Wotherspoon, Ayoma D. Attygalle, Hongxiang Liu, and Ming-Qing Du. Early detection of T-cell lymphoma with T follicular helper phenotype by RHOA mutation analysis. Haematologica. 2021; 106:xxx doi:10.3324/haematol.2020.265991

Publisher's Disclaimer. E-publishing ahead of print is increasingly important for the rapid dissemination of science. Haematologica is, therefore, E-publishing PDF files of an early version of manuscripts that have completed a regular peer review and have been accepted for publication. E-publishing of this PDF file has been approved by the authors. After having E-published Ahead of Print, manuscripts will then undergo technical and English editing, typesetting, proof correction and be presented for the authors' final approval; the final version of the manuscript will then appear in print on a regular issue of the journal. All legal disclaimers that apply to the journal also pertain to this production process. Early detection of T-cell lymphoma with T follicular helper phenotype by RHOA mutation analysis

Rachel Dobson,1 Peter Y Du,1 Lívia Rásó-Barnett,2 Wen-Qing Yao,1 Zi Chen,1 Calogero Casa,2 Hesham EI-Daly,2 Lorant Farkas,2,3 Elizabeth Soilleux,1,4 Penny Wright,4 John W Grant,4 Manuel Rodriguez-Justo,5 George A Follows,6 Hala Rashed,7 Margarete Fabre,8 E Joanna Baxter, 6 George Vassiliou,8Andrew Wotherspoon,9 Ayoma D. Attygalle,9 Hongxiang Liu,2 and Ming-Qing Du1,4

1Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK; 2The Haematopathology and Oncology Diagnostic Service, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; 3Department of Pathology, Akershus University Hospital, Lorenskog, Norway; 4Department of Histopathology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; 5Department of Pathology, Royal Free and University College Medical School, London, UK; 6Department of Haematology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; 7Department of Cellular Pathology, University Hospitals of Leicester, East Midlands Pathology Services, Leicester, UK; 8Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Department of Haematology, University of Cambridge, Cambridge, UK; 9Histopathology Department, The Royal Marsden Hospital, London, UK;

Author contributions: Experimental design, data collection and analysis: RD, PD, WQ, CC; Illumina sequencing analysis and variant calling: WQ, ZC; Case contribution and pathology review: LRB, HED, LF, ES, PW, JWG, MRJ, GAF, HR, AW, ADA, HL, MF, EJB, GV; Manuscript writing and preparation: MQD, RD; Research funding, study design and coordination: MQD. All authors commented on the manuscript and approve its submission for publication. The authors declare no conflict of interest

Acknowledgements: The authors would like to thank Shubha Anand and Yuanxue Huang for their assistance with using TapeStation, Graeme Clark and Ezequiel Martin for their assistance with Illumina sequencing and Fangtian Wu for carrying out DNA extraction on a control case.

Sources of Research support: The research in MQD's lab was supported by grants from Blood Cancer UK (13006, 15019), CRUK (C8333/A29707), and the Kay Kendall Leukaemia Fund (KKL582) UK. WY was supported by a research fellowship from the China Scholarship Council, and an International Collaborative Award from the Pathological Society of Great Britain and Ireland, UK. The Human Research Tissue Bank is supported by the NIHR Cambridge Biomedical Research Centre.

Running title: RHOA mutation analysis in early lymphoma diagnosis Key words: AITL, early diagnosis, RHOA mutation, qPCR

Correspondence to Professor Ming-Qing Du, Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge Box 231, Level 3, Lab Block, Addenbrooke’s Hospital, Hills Road, Cambridge, CB2 2QQ, United Kingdom Tel: +44 (0)1223 767092; Fax: +44 (0)1223 586670; Email: [email protected]

1

Word Count: Abstract: 249; Main manuscript text: 3898

Manuscript figure and tables: 4 figures and 1 table, 2 supplementary figures, 3 supplementary tables

2

ABSTRACT

Angioimmunoblastic T-cell lymphoma (AITL) and peripheral T-cell lymphoma with T follicular helper phenotype (PTCL-TFH) are a group of complex clinicopathological entities that originate from TFH cells and share a similar mutation profile. Their diagnosis is often a challenge, particularly at an early stage, due to a lack of specific histological and immunophenotypic features, paucity of neoplastic T cells and prominent polymorphous infiltrate. We investigated whether the lymphoma associated RHOA Gly17Val (c.50G>T) mutation, occurring in 60% of cases, is present in the early “reactive” lesions, and whether mutation analysis can help advance early lymphoma diagnosis. The RHOA mutation was detected by quantitative PCR with a locked nucleic acid (LNA) probe specific to the mutation, and a further PNA clamp oligonucleotide to suppress the amplification of the wild-type allele. The qPCR assay was highly sensitive and specific, detecting RHOA Gly17Val at an allele frequency of 0.03%, but not other changes in Gly17, nor in 61 controls. Among the 37 cases of AITL and PTCL-TFH investigated, RHOA Gly17Val was detected in 62.2% (23/37) of which 19 had multiple biopsies including preceding biopsies in 10 and follow up biopsies in 11 cases. RHOA Gly17Val was present in each of these preceding or follow up biopsies including 18 specimens that showed no evidence of lymphoma by combined histological, immunophenotypic and clonality analyses. The mutation was seen in biopsies 0-26.5 months (mean=7.87 months) prior to lymphoma diagnosis. Our results show that RHOA Gly17Val mutation analysis is valuable in the early detection of AITL and PTCL-TFH.

INTRODUCTION

Angioimmunoblastic T-cell lymphoma (AITL), peripheral T-cell lymphoma with T follicular helper phenotype (PTCL-TFH) and follicular T-cell lymphoma (FTCL) are a group of complex clinicopathological entities that originate from T follicular helper cells. These lymphomas are the most common among various T-cell malignancies in Western countries. Patients with these lymphomas commonly show an aggressive clinical course, with a median three year survival of only 30% (https://www.hmrn.org/statistics/disorders/34). The poor clinical outcome is largely due to a lack of understanding of their molecular mechanism and targeted therapy.

The diagnosis of these lymphoma entities, particularly at their early stage, is a challenge due to the lack of specific clinical and histological features, low tumor cell content and the presence of prominent polymorphous inflammatory infiltrates that often mask the neoplastic cells. To help the diagnosis, T-cell clonality analysis is commonly used, but this is frequently not helpful due to the paucity of malignant T cells. The diagnostic difficulty is further exacerbated by the increasing use of needle core biopsies to avoid more invasive surgical excision. When a lymphoproliferative lesion is suspected but uncertain on histological diagnosis, patients are commonly subjected to a “watch and wait” approach, and further biopsied when showing signs of progression. There is a wide range of genetic changes in T-cell lymphomas with TFH phenotype, including early TET2 and DNMT3A mutations which occur in haematopoietic stem cells, and late genetic changes which are specifically seen in lymphoma cells. 1-4 Among the latter, RHOA mutation is the most frequent,2,5-9 occurring in 60-70% of cases with Gly17Val (c.50G>T) accounting for 95% of the changes. Interestingly, mouse model studies have suggested that RHOA Gly17Val mutation induces TFH cell differentiation and, together with loss-of-function TET2 mutations, can promote AITL-like lymphoma development.10-12 Importantly, RHOA mutation is preferentially associated with the above lymphoma entities and has, so far, been shown only in the neoplastic T-cell population, but not in reactive B and T cells in these malignant conditions.2,5,13,14 Studies to date suggest that the RHOA Gly17Val mutation could be used as a marker for diagnosis of AITL and PTCL- TFH.6,15 However, it is unclear whether the mutation is present in tissue biopsies prior to AITL diagnosis and whether mutation analysis could help in the early detection of these lymphomas. To investigate this, we established a highly sensitive quantitative PCR assay to detect the RHOA mutation, and screened a large cohort (n=37 cases) of AITL and PTCL-TFH with multiple sequential biopsies together with 61 controls.

3

METHODS

Tissue materials and DNA extraction

The use of archival tissues for research was approved by the ethics committees of the involved institutions. In total, 78 tissue specimens from 37 patients (multiple consecutive specimens in 29 cases) with AITL (n=35) or PTCL-TFH (n=2), together with 61 controls including 13 reactive lymph nodes with paracortical expansion of T cells, 10 classical Hodgkin lymphoma and 1 marginal zone lymphoma with a prominent background of T cells and peripheral blood DNA samples from 16 individuals (TET2 & DNMT3A mutation in 2, TET2 mutation in 4, DNMT3A mutation in 5) with clonal hematopoiesis of indeterminate potential (CHIP) (1 individual classed as high-risk CHIP) were successfully investigated. DNA samples were prepared from a whole tissue section of each specimen. Purified DNA was obtained using QIAamp DNA-Micro kit (Qiagen), while crude DNA was obtained by digesting samples at 37°C overnight with proteinase K and NP-40 buffer. Purified DNA samples from 9 AITL or PTCL-TFH with known RHOA mutations [Gly17Val, c.50G>T, n=6 with variant allele frequency (VAF) ranging 2-32%; Gly17Leu, c.49-50GG>TT, n=2; Gly17Glu, c.50G>A, n=1; Ser26Arg, c.76A>C, n=1] were available from our previous study2, while crude DNA samples were available from routine clonality analysis or similarly prepared. The histology of these specimens were reviewed by specialist haematopathologists.

Quantitative PCR with peptide nucleic acid clamp and locked nucleic acid probe (PNA-LNA qPCR)

This was performed as previously described.16 The qPCR assay contained two probes and a PNA clamp (Figure 1A, Table S1). The total probe served as an internal control to monitor the PCR performance. The PNA clamp specifically and strongly binds to the wild-type DNA sequence, resists to the 5’ nuclease activity of Taq DNA polymerase, and thus blocks the wild-type allele from PCR amplification. This results in preferential amplification of the mutant allele which is detected specifically by the LNA mutant probe.

PCR primers with various probes were first tested using purified DNA samples, with the optimized conditions outlined below (Table S1). Briefly, the PNA-LNA PCR was carried out in a 20µl reaction containing 10µL Premix Ex Taq (Probe qPCR) Master Mix (Takara, Shiga, Japan), 0.2µM each forward and reverse primer, 0.1µM each total and mutant probe, 0.05µM PNA clamp probe, and 2µL of crude or 25ng purified DNA. Real time PCR was carried out in triplicate using Quantstudio 6 (Thermo Fisher Scientific, Waltham, MA, USA) with denaturation at 95°C for 30s followed by 45 cycles at 95°C for 3s, and 62°C for 30s.

Targeted sequencing using Fluidigm Access Array and Illumina MiSeq

This was performed on a selected case with consecutive tissue biopsies as described in our recent study.2 Each of the DNA samples were investigated in duplicate for mutations in TET2, DNMT3A, IDH2, RHOA, PLCG1, CCND3, CD28 and TNFRSF21 by Fluidigm PCR and Illumina MiSeq sequencing. Sequence reads alignment, variant calling, filtering to eliminate false positive and benign changes were carried out according to our previously established protocols.2 Only the reproducible variants that appeared in both replicates were regarded as a true change.

BaseScope in situ hybridization (ISH)

This was performed on a selected case. The sequence of the clonal TRB rearrangement in a case of AITL was available from a recent study.2 Based on the unique VDJ junctional sequence, unique BaseScope probes were designed and used to identify the lymphoma T cells by in situ hybridization. The BaseScope ISH was carried out according to the manufacturer's instructions (Advanced Cell Diagnostics, Newark, CA, USA).2 The ISH condition was optimized using the AITL specimen from which the clonal TRB rearrangement was sequenced, together with tonsils as a negative control.

4

RESULTS

PNA-LNA qPCR is highly sensitive and specific for RHOA Gly17Val (c.50G>T) detection

Upon the optimization of PCR conditions for RHOA mutation detection, we first determined the sensitivity of the qPCR assay using serial dilutions of three purified DNA samples with known VAF of Gly17Val (c.50G>T) into tonsil DNA. The PNA-LNA qPCR was highly sensitive, capable of detecting the mutation at 0.032% VAF (Figure 1B). As expected, the qPCR was highly specific to Gly17Val (c.50G>T) and showed no detectable signal of the LNA mutant probe with the DNA samples harboring other RHOA mutations including Gly17Leu (c.49-50GG>TT) and Gly17Glu (c.50G>A), nor tonsillar DNAs (Figure S1A).

As the crude DNA preparations were routinely used for clonality analysis in our clinical diagnostic laboratories, we tested whether such crude DNA samples were amenable for the above PNA-LNA qPCR (Figure S1C). Among the 10 crude DNA samples initially tested, 9 yielded excellent amplification comparable with the results from purified DNA samples. We then used crude DNA preparations for the qPCR in the remaining investigations, with high quality results from 139 of the 144 samples investigated. The 5 samples that failed to support the qPCR were essentially due to poor quality of DNA as evidenced by quality control PCR.

RHOA Gly17Val (c.50G>T) is detected in initial biopsies not diagnostic for lymphoma by conventional approach

To confirm that the RHOA mutation was lymphoma specific, not associated with reactive conditions proliferations or other lymphoproliferative disorders with enriched T cells, we investigated 45 tissue biopsies including 27 from lymph nodes, where T-cell clonality analysis was requested during routine histological diagnosis, but showed no evidence of a T-cell lymphoma. These included 13 specimens showing paracortical T-cell expansion or enriched CD4+ T cells, and 10 classic Hodgkin lymphomas with a prominent background of T cells. All these specimens were successful for the qPCR as indicated by the total probe control but were negative for the RHOA mutation. Similarly, we also showed absence of the RHOA mutation in peripheral blood (Figure S1D) from 16 individuals with CHIP (mutational information provided in Table S2), in keeping with the findings by whole exome or panel sequencing17,18.

We then investigated 37 cases of AITL (n=35) or nodal PTCL-TFH (n=2) with unknown RHOA mutation status, of which 29 had multiple longitudinal biopsies (2-5) available for RHOA analysis. RHOA mutation was seen in 23 cases, but was negative in the remaining 14 cases. Among the 23 cases with RHOA mutation, 19 cases had multiple biopsies. We reviewed the original histological diagnosis in each of these specimens and categorized them into three groups. Group-A specimens (n=26) showed clear evidence of lymphoma by histology and immunophenotype, further supported by clonal TCR rearrangement. Group-B (n=6) had suspicious histological and immunophenotypic findings, although not entirely diagnostic for lymphoma, which was ascertained by detection of strong clonal TCR gene rearrangements. Finally, group-C (n=18) were not diagnostic for lymphoma by combined histological, immunophenotypic and clonality analyses (Table S3). Interestingly, RHOA mutation was detected in each of these specimens, in the positive cases, including those not diagnostic for lymphoma by conventional integrated diagnostic investigations (Figure 2).

Characteristics of biopsies not diagnostic for lymphoma but RHOA mutation positive

Of the 23 cases of AITL or PTCL-TFH with RHOA mutation, the initial biopsy was not diagnostic in 10 cases including 2 with an excisional specimen. The time from the initial non-diagnostic biopsy to that establishing AITL or PTCL-TFH diagnosis ranged from 0 to 26.5 months, with an average of 7.87 months. All these initial non-diagnostic biopsies showed either polyclonal (n=4) or weak clonal (n=5) or oligoclonal TCR gene rearrangements (n=2) or failed (n=1) by BIOMED-2 TRB and TRG clonality analysis. 7 of these initial non-diagnostic biopsies were also subjected to BIOMED-2 B-cell clonality analysis and 6 showed a clonal IG

5 gene rearrangement. EBER in situ hybridization was carried out in 8 cases during routine diagnostic workout, and 4 showed variable positivity from a few scattered to numerous EBER positive cells, of which 3 displayed clonal IG gene rearrangement. In 4 cases, EBV-driven proliferation or classical Hodgkin lymphoma were considered in the initial diagnosis.

Six follow up biopsies were not diagnostic of involvement by AITL or PTCL-TFH by routine diagnostic investigations, and they included 2 and 2 skin specimens. In each specimen, a CD4+ T-cell infiltrate was noted, but a definite aberrant immunophenotype and expression of TFH markers could not be ascertained. T-cell clonality analysis showed weak polyclonal in 2 and a weak clonal or oligoclonal pattern in 2.

Representative cases

Case 30: A 79-year-old man presented with bilateral tender neck lymph nodes, and had mild sweats, but no weight loss or fever. Clinical examination revealed multiple bilaterally enlarged neck and groin lymph nodes (up to 1.5 cm diameter), and palpable liver and . Right level II neck lymph node excision biopsy showed partial effacement of the lymph node architecture and expansion of the interfollicular area by a polymorphous population of lymphoid cells, including B cells with plasmacytoid differentiation, and scattered large cells (Figure 3). These B cells expressed pan B-cell markers (CD20, CD79a, CD19), and MUM1, but were negative for CD10 and BCL6 (data not shown). A high proportion of the B cells were EBER positive and showed IG kappa light chain restriction. The lymphoid follicles appeared to be reactive and showed no apparent expansion of TFH cells, with only a few CD10 positive cells spilling out of the germinal centers (Figure 3). BIOMED-2 clonality analyses showed clonal IGH and IGK gene rearrangements, but a weak oligoclonal pattern with TRG and TRB. The histological diagnosis was uncertain: considering a clonal EBV positive polymorphous lymphoproliferation. Close follow up with a low threshold for re-biopsy was recommended.

Two months later, the patient presented with increasing fatigue, fever, maculopapular chest rash and increased size of peripheral lymphadenopathy. Positron-emission tomography (PET) scan revealed extensive bilateral cervical, mediastinal, bilateral iliac and groin lymphadenopathy. Left level V neck lymph node excision biopsy showed partial effacement of the lymph node architecture by an infiltrate of medium sized atypical lymphoid cells with regressed follicles (Figure 3). There was hyperplasia of follicular (FDC) meshworks and high endothelial venules with the atypical lymphoid cells clustered in their vicinity. The atypical lymphoid cells were positive for CD3, CD5 and TFH markers (PD1, CD10, ICOS, BCL6) (Figure 3). EBER in situ hybridization revealed only scattered positive cells. BIOMED-2 clonality analyses showed clonal TRB and TRG gene rearrangements, and also weak clonal IGH and IGK gene rearrangements which were different from the previous biopsy in the size of their amplified IG products. A diagnosis of AITL was made. The patient was initially treated with 6 cycles of CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisone), then Avelumab (cycle 12 at the most recent follow up) under the AVAIL-T trial, and was well, showing no constitutional symptoms or palpable cervical lymph nodes, 20 months following AITL diagnosis.

Retrospective analysis showed the presence of RHOA Gly17Val mutation in the above two specimens by qPCR and targeted sequencing with 3% and 23% VAF in the first and second biopsy, respectively. The targeted sequencing also revealed a pathogenic nonsense substitution in DNMT3A (c.2311C>T, p.R771*) in the first and second biopsy at 17% and 26% VAF, respectively.

Case 7: An 82-year-old man, with remission of a previous mantle cell lymphoma, presented with skin rash on the right calf. A punch biopsy showed a mild perivascular infiltrate by CD3+ T cells, with a histological diagnosis of panniculitis (Figures 4 & Table 1). To investigate potential lymphoma relapse, lymph node core biopsies were taken at 11 and 12 month follow up, and both specimens showed no evidence of mantle cell lymphoma, but revealed cardinal features of AITL and also a prominent pleomorphic infiltrate including an EBER-positive B-cell population with HRS-like morphology and immunophenotype (Figures 4 & Table 1). T-

6 cell clonality analysis demonstrated clonal rearrangements by TRG-A and TRB-A with identical sized amplified products between the two biopsies, and an additional clonal rearrangement by TRB-C in the second biopsy. Further follow up biopsies were taken at 25 and 26 month follow up, and both biopsies showed no apparent evidence of AITL, but a polymorphous infiltrate with a more prominent EBER-positive B-cell population that had HRS cell morphology and immunophenotype (Figures 4 & Table 1). Both specimens showed no evidence of the clonal TRG/TRB rearrangements seen in the early lymph node biopsies, although the fifth biopsy displayed an isolated clonal rearrangement by TRB-C. B-cell clonality analyses demonstrated polyclonal IG gene rearrangements in both specimens (Figures 4 & Table 1). A classical Hodgkin lymphoma arising from the EBV positive B-cell component of the AITL was considered.

In a retrospective study, the TRB-A and B PCR products from the second biopsy were sequenced using Illumina MiSeq platform and a dominant TRBV5-J2 rearrangement (86%) was identified.2 Based on the unique VDJ junctional sequence, we designed unique BaseScope probes to identify the lymphoma T cells by in situ hybridization (Figure 4 & Figure S2). As expected, both the second and third biopsies with AITL diagnosis showed diffuse positivity with the lymphoma clone-specific probe. Interestingly, the initial skin biopsy also displayed isolated positive cells, while the fifth biopsy with EBV-LPD diagnosis yielded a negative result. Both the second (AITL) and fifth (EBV-LPD) biopsies were investigated by panel sequencing for recurrent somatic mutations, and this identified 5 shared mutations including 1 DNMT3A, 3 TET2 and 1 RHOA changes between the two specimens, and 1 further TET2 mutation only in the second specimen (Table 1). In general, the mutation load in each of the above shared changes were much higher in the second biopsy (AITL) than the fifth biopsy (EBV-LPD). This was particularly striking for the RHOA Gly17Val mutation, with 20% VAF in the second but only 1% VAF in the fifth biopsy. As expected, qPCR for the RHOA mutation demonstrated its strong positivity in both the second and third biopsies showing AITL, but a weak positive signal in the initial skin biopsy, and the fourth and fifth biopsies displaying classical Hodgkin lymphoma-like EBV-LPDs (Figure 4 & Figure S2).

DISCUSSION

By using a highly sensitive qPCR assay, we confirm that RHOA Gly17Val (c.50G>T) mutation is specifically associated with AITL or PTCL-TFH, but not found in other lymphoproliferative conditions including those with infiltration of florid T-helper cells. More importantly, detection of RHOA Gly17Val (c.50G>T) mutation could help early detection of AITL and PTCL-TFH, and also diagnosis of their extranodal involvement.

The difficulty for histological diagnosis of AITL/PTCL-TFH is well recognized, particularly when the neoplastic cell content is low, inconspicuous by histological and immunophenotypic assessment and undetectable by analysis of TCR gene rearrangements. Apart from the paucity of neoplastic T cells, the polymorphous infiltrate, particularly the presence of numerous EBV-positive B cells, including Reed-Sternberg-like cells (EBV-positive or negative), may lead to diagnostic consideration of EBV-driven B-cell proliferation.19-21 Such misleading diagnostic features may also be reinforced by the frequent demonstration of clonal IG gene rearrangement. As shown in this study, EBV-driven B-cell proliferation was the diagnostic consideration in 4/11 of the initial biopsies. Indeed, it has been reported by several independent studies that EBV-associated lymphoproliferation is a frequent pitfall in AITL diagnosis.19-22

In the present study, core biopsy accounts for the majority of the initial specimens that are not diagnostic. It is possible that these core biopsies are not totally representative, with characteristic lymphoma components missed due to sampling error. Of note, two initial non-diagnostic biopsies are excisional lymph node specimens, indicating that an absence of diagnostic features of AITL/PTCL-TFH is also a real issue in the initial biopsies. Interestingly, the time interval between the initial non-diagnostic and the follow up biopsies that established the diagnosis varies considerably, ranging from 0 to 26.5 months. In the cases with a long interval, it is likely that the initial biopsy represents an early premalignant lesion, while the follow up biopsy reflects more progressed disease, thus having more cardinal features for AITL/PTCL-TFH diagnosis. While in

7 the cases with a short interval, it is possible that the enlarged lymph nodes are variably involved by AITL/PTCL-TFH and the initial non-diagnostic biopsies represent early involvement by the lymphoma.

Both the above possibilities may exist, without excluding the other. It is impossible to distinguish the two scenarios based on analysis of a single biopsy including RHOA mutation analysis. Nonetheless, detection of RHOA Gly17Val (c.50G>T) mutation can certainly raise the alarm for more in-depth histological and immunophenotypic investigations, for example more careful search for evidence of TFH cell expansion as shown in the first biopsy in case 30. It is important to emphasize that a positive detection of RHOA mutation is not equal to lymphoma diagnosis as the mutational analysis by qPCR or targeted sequencing is highly sensitive, and the mutation is seen in biopsies without histological evidence of AITL. As discussed above, the RHOA mutation positive non-diagnostic biopsy may represent a premalignant lesion or an early involvement by the lymphoma. Therefore, the RHOA mutation analysis should be used as an auxiliary tool with the results interpreted in the context of histological and immunophenotypic findings. If a histological diagnosis of AITL/PTCL-TFH cannot be made, an excision biopsy or a low threshold for an early follow up biopsy, where appropriate, is indicated.

In a few case studies, RHOA mutation was detected in circulating free DNA (cfDNA) or peripheral blood lymphocytes from patients with AITL/PTCL-TFH,15 and the mutation burden appeared to correlate with the treatment outcome.23,24 It remains to be investigated whether the mutation is detectable in cfDNA or peripheral blood lymphocytes at the time of initial non-diagnostic biopsies. Given that the RHOA mutation burden in cfDNA and peripheral blood lymphocytes reflects, at least theoretically, the overall lymphoma load, simultaneously analyzing blood samples, in addition to tissue biopsy, could add further value in lymphoma diagnosis, particularly when the specimen is not representative.

Apart from the initial biopsies, the diagnosis of extranodal involvement by AITL/PTCL-TFH such as skin and bone marrow is also difficult due to similar reasons to those discussed above, including low tumor cell content and polymorphous infiltrate.25 In particular, extranodal infiltrates may not harbor the cardinal features of AITL such as TFH marker expression in the neoplastic T cells and FDC meshworks. The DNA sample from these extranodal sites is often not informative for clonality analysis due to insufficient lymphoid cells and/or poor DNA quality. As shown in our study, RHOA mutation analysis is highly valuable in confirmation of AITL/PTCL-TFH involvement in follow up biopsies. Similarly, RHOA mutation analysis is valuable in diagnosis of AITL/PTCL-TFH from cytological specimens.9

Aside from the RHOA mutation, there are several other genetic changes including IDH2, CD28, PLCG1, VAV1 and TNFRSF21 mutations, and VAV1-STAP2, CTLA4-CD28 and ITK-SYK fusion, which occur at variable frequencies in AITL and PTCL-TFH.2,26-28 Of note, mutations in VAV1, a signaling molecule downstream of TCR, occurs in 8.2% of AITL and appears to be mutually exclusive from the RHOA mutation.28 Detection of these additional lymphoma associated genetic changes could also help the early detection of AITL/PTCL-TFH, together with RHOA mutation detection, potentially being valuable for diagnosis in up to 90% of these T-cell lymphomas. These diverse genetic changes could be readily investigated by targeted sequencing either alone or as part of a comprehensive lymphoma panel.

The detailed analyses of multiple biopsies in case 7 also reveal the mutation burden in non-malignant T cells. The fifth lymph node biopsy shows little involvement by AITL as the lymphoma clone is undetectable by BaseScope ISH and the RHOA mutation is only present at 1% VAF, but the specimen bears a high TET2 mutation burden (20% VAF). This implies that the TET2 mutation must be present in a high proportion of reactive B and T cells,1,5,29-31 probably up to 40% of the total cell population, well above the EBER-positive cell fraction (~5%). Hence, this enlarged lymph node is essentially caused by lymphoid proliferation driven by TET2 mutation and/or EBV infection. These findings further highlight the markedly variable histological presentation of enlarged lymph nodes in patients with AITL and, hence, the potential sampling error in AITL diagnosis. In this context, it is pertinent to investigate RHOA and other lymphoma associated mutations in

8 patients with advanced age and biopsies showing EBV positive polymorphous infiltrates, to explore how such mutation analyses can help circumventing this diagnostic pitfall. At the genetic level, the high number of TET2 mutations and their high presence in non-neoplastic T cells suggest that the patient most likely had an underlying CHIP with TET2 and DNMT3A mutations that occurred in haematopoietic stem cells, consequently extending to their progenies.

In general, patients with AITL typically show an aggressive clinical course and respond poorly to the currently available therapies. In contrast, case 7 appeared to be exceptional, showing a slow disease progression in the absence of any treatment, although it is not possible to rule out a response to R-GCVP that aimed to treat the EBV-associated proliferations (Table 1). An indolent clinical course has been previously reported for some patients with AITL showing pattern-1 histology, including those treated only with steroids.22,32 It remains to be investigated how to identify such indolent cases at the time of diagnosis and stratify their clinical management accordingly, particularly in view of the advance in early detection of AITL.

In summary, we have shown that RHOA mutation is specifically associated with AITL and PTCL-TFH and their related lesions, and investigation of the mutation is highly valuable in early detection of these T-cell lymphomas and their extranodal involvement.

REFERENCES

1. Nguyen TB, Sakata-Yanagimoto M, Asabe Y, et al. Identification of cell-type-specific mutations in nodal T-cell lymphomas. Blood Cancer J. 2017;7(1):e516.

2. Yao WQ, Wu F, Zhang W, et al. Angioimmunoblastic T-cell lymphoma contains multiple clonal T-cell populations derived from a common TET2 mutant progenitor cell. J Pathol. 2020;250(3):346-357.

3. Iqbal J, Amador C, McKeithan TW, Chan WC. Molecular and Genomic Landscape of Peripheral T-Cell Lymphoma. Cancer Treat Res. 2019;176:31-68.

4. Xie M, Lu C, Wang J, et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med. 2014;20(12):1472-1478.

5. Sakata-Yanagimoto M, Enami T, Yoshida K, et al. Somatic RHOA mutation in angioimmunoblastic lymphoma. Nat Genet. 2014;46(2):171-175.

6. Nakamoto-Matsubara R, Sakata-Yanagimoto M, Enami T, et al. Detection of the G17V RHOA mutation in angioimmunoblastic T-cell lymphoma and related lymphomas using quantitative allele-specific PCR. PLoS One. 2014;9(10):e109714.

7. Palomero T, Couronne L, Khiabanian H, et al. Recurrent mutations in epigenetic regulators, RHOA and FYN kinase in peripheral T cell lymphomas. Nat Genet. 2014;46(2):166-170.

8. Ondrejka SL, Grzywacz B, Bodo J, et al. Angioimmunoblastic T-cell Lymphomas With the RHOA p.Gly17Val Mutation Have Classic Clinical and Pathologic Features. Am J Surg Pathol. 2016;40(3):335-341.

9. Lee PH, Weng SW, Liu TT, et al. RHOA G17V mutation in angioimmunoblastic T-cell lymphoma: A potential biomarker for cytological assessment. Exp Mol Pathol. 2019;110:104294.

9

10. Zang S, Li J, Yang H, et al. Mutations in 5-methylcytosine oxidase TET2 and RhoA cooperatively disrupt T cell homeostasis. J Clin Invest. 2017;127(8):2998-3012.

11. Cortes JR, Ambesi-Impiombato A, Couronné L, et al. RHOA G17V Induces T Follicular Helper Cell Specification and Promotes Lymphomagenesis. Cancer Cell. 2018;33(2):259-273.

12. Ng SY, Brown L, Stevenson K, et al. RhoA G17V is sufficient to induce and promotes T- cell lymphomagenesis in mice. Blood. 2018;132(9):935-947.

13. Dobay MP, Lemonnier F, Missiaglia E, et al. Integrative clinicopathological and molecular analyses of angioimmunoblastic T-cell lymphoma and other nodal lymphomas of follicular helper T-cell origin. Haematologica. 2017;102(4):e148-e151.

14. Yoo HY, Sung MK, Lee SH, et al. A recurrent inactivating mutation in RHOA GTPase in angioimmunoblastic T cell lymphoma. Nat Genet. 2014;46(4):371-375.

15. Hayashida M, Maekawa F, Chagi Y, et al. Combination of multicolor flow cytometry for circulating lymphoma cells and tests for the RHOA(G17V) and IDH2(R172) hot-spot mutations in -free DNA as liquid biopsy for the diagnosis of angioimmunoblastic T-cell lymphoma. Leuk Lymphoma. 2020;61(10):2389- 2398.

16. Tanzima Nuhat S, Sakata-Yanagimoto M, Komori D, et al. Droplet digital polymerase chain reaction assay and peptide nucleic acid-locked nucleic acid clamp method for RHOA mutation detection in angioimmunoblastic T-cell lymphoma. Cancer Sci. 2018;109(5):1682-1689.

17. Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med. 2014;371(26):2488-2498.

18. Lewis NE, Petrova-Drus K, Huet S, et al. Clonal hematopoiesis in angioimmunoblastic T-cell lymphoma with divergent evolution to myeloid neoplasms. Blood Adv. 2020;4(10):2261-2271.

19. Steciuk MR, Massengill S, Banks PM. In immunocompromised patients, Epstein-Barr lymphadenitis can mimic angioimmunoblastic T-cell lymphoma morphologically, immunophenotypically, and genetically: a case report and review of the literature. Hum Pathol. 2012;43(1):127-133.

20. Nicolae A, Pittaluga S, Venkataraman G, et al. Peripheral T-cell lymphomas of follicular T-helper cell derivation with Hodgkin/Reed-Sternberg cells of B-cell lineage: both EBV-positive and EBV-negative variants exist. Am J Surg Pathol. 2013;37(6):816-826.

21. Laforga JB, Gasent JM, Vaquero M. Potential misdiagnosis of angioimmunoblastic T-cell lymphoma with Hodgkin's lymphoma: a case report. Acta Cytol. 2010;54(5 Suppl):840-844.

22. Tan LH, Tan SY, Tang T, et al. Angioimmunoblastic T-cell lymphoma with hyperplastic germinal centres (pattern 1) shows superior survival to patterns 2 and 3: a meta-analysis of 56 cases. Histopathology. 2012;60(4):570-585.

23. Sakata-Yanagimoto M, Nakamoto-Matsubara R, Komori D, et al. Detection of the circulating tumor DNAs in angioimmunoblastic T- cell lymphoma. Ann Hematol. 2017;96(9):1471-1475.

24. Nguyen TB, Sakata-Yanagimoto M, Fujisawa M, et al. Dasatinib Is an Effective Treatment for Angioimmunoblastic T-cell Lymphoma. Cancer Res. 2020;80(9):1875-1884.

10

25. Attygalle AD, Diss TC, Munson P, Isaacson PG, Du MQ, Dogan A. CD10 expression in extranodal dissemination of angioimmunoblastic T-cell lymphoma. Am J Surg Pathol. 2004;28(1):54-61.

26. Vallois D, Dobay MP, Morin RD, et al. Activating mutations in genes related to TCR signaling in angioimmunoblastic and other follicular helper T-cell-derived lymphomas. Blood. 2016;128(11):1490-1502.

27. Rohr J, Guo S, Huo J, et al. Recurrent activating mutations of CD28 in peripheral T-cell lymphomas. Leukemia. 2016;30(5):1062-1070.

28. Fujisawa M, Sakata-Yanagimoto M, Nishizawa S, et al. Activation of RHOA-VAV1 signaling in angioimmunoblastic T-cell lymphoma. Leukemia. 2018;32(3):694-702.

29. Couronné L, Bastard C, Bernard OA. TET2 and DNMT3A mutations in human T-cell lymphoma. N Engl J Med. 2012;366(1):95-96.

30. Quivoron C, Couronné L, Della Valle V, et al. TET2 inactivation results in pleiotropic hematopoietic abnormalities in mouse and is a recurrent event during human lymphomagenesis. Cancer Cell. 2011;20(1):25-38.

31. Schwartz FH, Cai Q, Fellmann E, et al. TET2 mutations in B cells of patients affected by angioimmunoblastic T-cell lymphoma. J Pathol. 2017;242(2):129-133.

32. Ch'ang HJ, Su IJ, Chen CL, et al. Angioimmunoblastic lymphadenopathy with dysproteinemia--lack of a prognostic value of clear cell morphology. Oncology. 1997;54(3):193-198.

11

TABLES: Table 1. Summary of histological, immunohistochemical, clonality analysis and genetic findings in case 7.

First biopsy Second biopsy Third biopsy Fourth biopsy Fifth biopsy

st Time after the 1 biopsy 11 months later 11.5 months later 33 months later 35 months later Episode of non-specific symptoms; Skin rash, fevers; Slowing enlarging lymph node with increasing Clinical lymphadenopathy revealed by CT, but All self-resolved with no symptoms. Treated with 6 cycles of R-GCVP*and presentation relatively low PET signal; opted for close therapy achieved CR; Ongoing remission in May 2020 surveillance. Lymph node core Left axillary lymph Right axillary lymph Right axillary lymph node Biopsy site Skin punch biopsy biopsy node core biopsy node core biopsy core biopsy

nd Lymph node structure Predominant infiltrate Very similar to the 2 effaced by Normal structure largely of small to medium biopsy. polymorphous effaced by polymorphous sized T cells with Predominantly small infiltrate with infiltrate with prominent vasculocentric pattern, to medium sized T scattered large atypical Mild perivascular large atypical cells Histology CD4+, some CD10+ and cells associated with cells expressing CD30, infiltrate, expressing CD30, CD20, & BCL6+ T cells possibly HEV proliferation, CD20 (weak), CD79, predominantly CD3+ T + + CD79a, PAX5, MUM1, Immuno- spilled outside B-cell CD4 , PD1 , BCL6 PAX5, BCL6, MUM1, cells, admixed + BCL6(weak), and EBER+, phenotype follicle, expanded FDC variable ; OCT2, BOB1, and and B cells but not CD15, CD10, meshworks; Expanded FDC and EBER+, but not CD15. CYCLIN D1, ALK and CD25. Scattered large EBER+ pleomorphic infiltrate, CD3 T cells: largely There are no detectable T B cells: CD30+, CD15 Scattered large B cells CD4+, some ICOS+, + with HRS morphology + cell abnormalities weak + + occasional PD1 , but CD30 , CD15 weak - - CD10 and CXCL13

Diagnosis Panniculitis AITL AITL Classical HL Classical HL

T- cell clonality

TRG-A n/a 217bp 217bp Poly Poly

TRG-B n/a Poly Poly Poly Poly TRB-A n/a 245bp 245bp Poly Poly TRB-B n/a Poly Poly Poly Poly TRB-C n/a 302bp Poly Poly 187bp

B-cell clonality n/a n/a n/a Poly Poly

BaseScope- ISH Few positive cells Diffuse positive Diffuse positive n/a Negative for TRB-V5- DNMT3A (VAF: 34%) DNMT3A (VAF:20%) (c.920C>T; p.P307L) (c.920C>T; p.P307L) TET2 (VAF: 8%) TET2 (VAF: 4%) (c.3646C>T; p.R1216X) (c.3646C>T; p.R1216X) TET2 (VAF: 36%) TET2 (VAF: 20%) (c.3781C>A; pR1261S) (c.3781C>A, pR1261S) Targeted n/a TET2 (VAF: 3%) n/a n/a TET2 (VAF: 8%) Sequencing (c.3866G>T; p.C1289F) (c.3866G>T; p.C1289F)

TET2 (VAF: 18%) (c.4947T>A; p.Y1649X)

RHOA (VAF: 20%) RHOA (VAF: 1%) (c. 50G>T; p.G17V) (c.50G>T; p.G17V)

RHOA c.50G>T Weak positive Strong positive Strong positive Weak positive Weak positive n/a: not available; R-GCVD: Rituximab, Gemcitabine, Cyclophosphamide, Vincristine, Prednisolone; CR: complete remission; VAF: variant allele frequency

12

FIGURE LEGENDS:

Figure 1. Detection of RHOA p.Gly17Val (c.50G>T) by real time PCR with PNA/LNA probes.

A) Schematic illustration of the real time PCR design. The total probe is used as an internal control to monitor the PCR performance. The PNA (peptide nucleic acid) clamp binds to the wild-type sequence, resists the 5’ nuclease activity of Taq DNA polymerase and thus blocks the wild-type allele from PCR amplification. This results in preferential amplification of the mutant allele which is detected specifically by the LNA (locked nucleic acid) mutant probe.

B) The PCR assay is highly sensitive, capable of detecting the RHOA mutation at 0.03% variant allele frequency (VAF) based on serial dilutions of an AITL sample with known mutation allele frequency by next generation sequencing (left panel). For simplicity, only the LNA mutant but not total probe signals are shown. The PCR assay shows a linear correlation among the serial dilutions (right panel).

Figure 2. Analysis of RHOA (c.50G>T; p.Gly17Val) mutation by qPCR in the RHOA positive cases with longitudinal biopsies in patients with AITL (only including cases with at least one biopsy classed as not diagnostic (“group B” or “group C”)).

The diagnosis of these specimens were reviewed, and categorized into three groups: Group A (filled circle): lymphoma diagnosed by histology and immunophenotype, further supported by clonal TCR gene rearrangement; Group B (half-filled circle): lymphoma not diagnostic by histology and immunophenotype alone, but ascertained by clonal TCR gene rearrangement; Group C (open circle): lymphoma not diagnosed by combined analyses. Red (regardless of the symbol: Including outline only, half-filled, and completely filled symbols) indicates RHOA G17V mutant positive biopsy. Grey indicates RHOA G17V status unknown. E denotes extranodal biopsies. * denotes lymph node excision biopsies and all others are core biopsy specimens. Open square denotes diagnosis of classical Hodgkin lymphoma (with no apparent evidence of AITL). Case 38 lacks a final diagnosis as the patient passed away, thus indicated by a dashed line. Cases with only Group A multiple biopsies are not included in this figure.

Figure 3. Histological and immunophenotypic findings in case 30.

The first lymph node excision biopsy shows a partial effacement of the lymph node architecture by polymorphous infiltrates, particularly B cells with plasmacytoid differentiation. A high proportion of B cells were EBER positive and showed IG kappa light chain restriction (not shown). The lymphoid follicles appear to be reactive and showed no apparent expansion of TFH cells, with only few CD10 positive cells spilling out of the , consistent with pattern 1 histology of AITL.

The second lymph node biopsy shows effacement of the lymph node architecture by medium sized atypical lymphoid cells with regressed follicles. There is a prominent proliferation of follicular dendritic cell (FDC) meshworks and high endothelial venules with the atypical lymphoid cells clustered in their vicinity. The atypical lymphoid cells are T cells expressing TFH markers, spilling out of the germinal center to the interfollicular region. EBER in situ hybridization shows only scattered positive cells.

Figure 4. Histological and immunophenotypic findings in case 7.

The first biopsy shows a subcutaneous perivascular infiltrate of CD3+ T cells with vasculitic features. The third biopsy shows cardinal features of AITL and also a prominent pleomorphic infiltrate including an EBER- positive B-cell population with HRS-like morphology and immunophenotype.

13

The fifth biopsy shows no apparent evidence of AITL, but a polymorphous infiltrate with a more prominent EBER-positive B-cell population that had HRS cell morphology and immunophenotype, rosetting by CD4+ T cells.

14

SUPPLEMENTARY TABLES

Table S1: Sequences of PCR primers and probes used for RHOA mutation detection.

Sequence (5’-3’) Manufacturer

Forward primer CCTATGACTTCTTGTGCATTGC Sigma-Aldrich

Reverse primer TACACCTCTGGGAACTGGTC Sigma-Aldrich

Total probe [Cyanine5]GCTGCCATCCGGAAGAAACTG[BHQ3] Sigma-Aldrich (Cy5)

Panagene PNA wild-type (Cambridge AGCCTGTGGAAAGACA clamp Research Biochemicals)

LNA mutant [6FAM]AGCC[+T]GT[+G][+T][+A]AAGA[+C]A[BHQ1] Sigma-Aldrich probe (Fam)

Table S2: Mutational information for clonal hematopoiesis of indeterminate potential (CHIP) samples.

Sample RHOA Gene Nucleotide Amino acid VAF qPCR change change (%)

CHIP 1 Negative SRSF2 c.C284A p.P95H 7.4

CHIP 2 Negative JAK2 c.G1849T p.V617F 14.7

TET2 c.A3797G p.N1266S 2.9

CHIP 3 Negative TET2 c.C2020T p.Q674X 17.5

c.G3893A p.C1298Y 15.8

c.G4249T p.V1417F 3.1

TP53 c.C523G p.R175G 3.5

CHIP 4 Negative DNMT3A c.1154delC p.P385fs 9.1

SF3B1 c.A2098G p.K700E 10.4

c.G1874T p.R625L 6.5

CHIP 5 Negative CBL c.2434+1G>A Essential splicing site 1.1

DNMT3A c.2173+1G>A Essential splicing site 26.1

PPM1D c.C1570T p.Q524X 4.8

CHIP 6 Negative CBL c.G570A p.W190X 2.3

SF3B1 c.A2098G p.K700E 0.9

SF3B1 c.G1998C p.K666N 34.2

CHIP 7 Negative DNMT3A c.2086delC p.Q696fs 0.9

TP53 c.G818A p.R273H 1.4

U2AF1 c.A470C p.Q157P 11.8

CHIP 8 Negative DNMT3A c.2567delA p.E856fs 5.4

c.1122+1G>- Essential splicing site 1.3

TET2 c.C863G p.P288R 1.9

CHIP 9 Negative DNMT3A c.G1904A p.R635Q 4.0

TP53 c.G1006T p.E336X 1.9

CHIP 10 Negative SF3B1 c.G1998T p.K666N 10.6

U2AF1 c.A470G p.Q157R 10.8

CHIP 11 Negative DNMT3A c.G2645A p.R882H 32.1

CHIP 12 Negative GNB1 c.A169G p.K57E 4.2

SF3B1 c.G1998T p.K666N 16.2

TET2 c.944delC p.S315fs 3.2

TET2 c.1305delC p.H435fs 1.2

TET2 c.A3821G p.Q1274R 3.3 TET2 c.T3965A p.L1322Q 3.4

TET2 c.C4624T p.Q1542X 1.3

TET2 c.C5473T p.Q1825X 40.9

CHIP 13 Negative SRSF2 c.C284T p.P95L 11.7

CHIP 14 Negative DNMT3A c.G989A p.W330X 6.5

SRSF2 c.C284A p.P95H 4.0

TET2 c.C3116A p.S1039X 2.9

c.4228delC p.P1410fs 2.3

c.4931_4935del p.P1644fs 1.6

CHIP 15 Negative SF3B1 c.2098A>G p.K700E 40.4

(High-risk) CHIP 16 Negative TET2 c.2031_2032delTG Cys677fs 36.4

Table S3: Summary of laboratory data of non-diagnostic biopsies in AITL with RHOA p.Gly17Val

mutation.

biopsy

-

for re for

cell phenotype, phenotype, cell

-

cell lymphoma cell

-

advised advised

cell lymphoma, but but lymphoma, cell

-

cell lymphoma cell

diagnosis

-

diagnostic

cell content and no evidence evidence no and content cell

clonality analysis clonality

by a T a by

Final histological opinion opinion histological Final

cell lymphoma, although shown by by shown although lymphoma, cell

-

suspicious but not diagnostic of diagnostic not but suspicious

lesional

cell component render difficulty in difficulty render cell component

-

involvement T a by involvement

lacking convincing histological and histological convincing lacking

lack of molecular evidence, thus not not thus evidence, molecular of lack

by AITL, but low number of assumed assumed of number low but AITL, by

B

Suspicious T a for Suspicious

No definite T aberrant definite No

Suspicious lymphoma involvement, but involvement, lymphoma Suspicious

of clonal TCR gene rearrangement, thus thus rearrangement, gene TCR clonal of

TCR rearrangement, thus not diagnostic not thus rearrangement, TCR

No histological evidence of involvement involvement of evidence histological No

No histological evidence of involvement involvement of evidence histological No

Suspicious of bone marrow involvement involvement marrow bone of Suspicious

by clonality and flow cytometry analyses analyses cytometry flow and clonality by

Panniculitis (please also refer to Table 1) Table to refer also (please Panniculitis

Little tissue for evaluation and dominant dominant and evaluation for tissue Little

not diagnostic as no supporting evidence evidence supporting no as diagnostic not

neoplastic cells and no evidence of clonal clonal of evidence no and cells neoplastic

by a T a by

Low Low

molecular evidence, evidence, molecular

cell cell

-

n/a

n/a

n/a

n/a

n/a

n/a

B

Clonal

clonality

Polyclonal

n/a

AITL

Weak Weak

Weak Weak

Clonal, Clonal,

identical identical

polyclonal

Polyclonal

polyclonal polyclonal

Polyclonal

polyclonal polyclonal

cell clonality cell

of previous previous of

background

Weak clonal Weak

Weak clonal Weak

-

Weak clonal in clonal Weak

T

pattern to that that to pattern

biopsy showing biopsy

T T

+

cell cell

ISH

-

-

cell cell

-

EBER

and CD8 and

negative

+

cell markers, markers, cell

-

paratrabecular

ISH

paratrabecular

-

, and plasma cells. cells. plasma and ,

cell markers cell

-

EBER

cell markers. markers. cell

-

, expressing pan T pan expressing ,

T cells, but very few few very but cells, T

+

+

immunoblasts

T cells, showing possible of loss possible showing cells, T

+

+

immunoblasts

immunoblasts

, no evidence of loss of pan T pan of loss of no evidence ,

+

+

See Table 1 Table See

markers

negative

histiocytes

, no loss of pan T pan of no loss ,

+

expressing PD1 expressing

parenchyma and portal tracts portal and parenchyma

Occasional EBER Occasional

high number of EBER of number high

, comprising sheets of histiocytes, in areas forming forming areas in histiocytes, of , sheets comprising

Histological & immunohistochemical findings findings immunohistochemical & Histological

cells, predominantly CD4 predominantly cells,

cells with no evidence T pan of loss of evidence no with cells

-

specific changes. A focal cluster of multiple perivascular perivascular multiple of cluster focal A changes. specific

-

Small, fragmented and poorly fixed tissue cores showing showing cores tissue fixed poorly and fragmented Small,

lymphoid aggregates, dominantly CD4 dominantly aggregates, lymphoid

markers, with a small subset PD1 positive. positive. PD1 subset small a with markers,

Non

aggregates of histiocytes, numerous small to slightly enlarged enlarged slightly to small numerous histiocytes, of aggregates

Mild perivascular chronic inflammatory cell infiltration, a plus cell infiltration, inflammatory chronic Mild perivascular

periadnexal

location, which are composed of small mixed B and T cells. The cells. T and B mixed small of composed are which location,

CD7, occasionally PD1 positive, but negative for CD10 and BCL6. BCL6. and CD10 for negative but positive, PD1 occasionally CD7,

expansion. T cells are predominantly CD4 predominantly are cells T expansion.

There are numerous lymphoid aggregates, with a a with aggregates, lymphoid numerous are There

Focal inflammatory cells including lymphocytes and histiocytes in histiocytes and lymphocytes including cells inflammatory Focal

immunohistochemistry. No evidence T pan of loss of evidence No immunohistochemistry.

There are no recognizable follicular structures on morphology and and on morphology structures follicular recognizable no are There

There is a nodular and interstitial infiltrate of small lymphoid cells, cells, lymphoid small of infiltrate interstitial and nodular a is There

granulomas with occasional giant cells, and inflammatory cells and and cells inflammatory and cells, giant occasional with granulomas

majority of T cells CD4 are cells T of majority

There are multiple intraparenchymal & occasionally occasionally & intraparenchymal multiple are There

lymphocytes. Immunohistochemistry shows mixed CD4 mixed shows Immunohistochemistry lymphocytes.

small lymphoid cells, CD30 positive positive CD30 cells, lymphoid small

mainly T mainly

of the dermis and also the subcutis. The infiltrate is perivascular and perivascular is infiltrate The subcutis. the also and dermis the of

There is a nodular dermal inflammatory infiltrate, involving levels all involving infiltrate, inflammatory dermal nodular a is There

Small and poorly preserved tissue fragments. A mixed population population of mixed A fragments. tissue preserved poorly and Small

predominantly lobular panniculitis associated with focal fat necrosis. necrosis. fat focal with associated panniculitis lobular predominantly

The infiltrate is predominantly small T cells, admixed with B cells and and cells B with admixed cells, T small predominantly is infiltrate The

polymorphous infiltrates, particularly histiocytes and T cells, FDC and cells, Tand histiocytes particularly infiltrates, polymorphous

lymphoid cells which are mainly CD4 mainly are which cells lymphoid

C

C

C

C

C

C

C

C

C

category

Diagnostic Diagnostic

biopsy

biopsy

follow up up follow

Follow up Follow

Follow up Follow

Follow up Follow

Preceding

Preceding

Preceding

Preceding

Preceding

diagnostic diagnostic

(Preceding) (Preceding)

Preceding or or Preceding

Same date as as date Same

Core Core

Core Core

Core Core

Punch Punch

Punch Punch

biopsy

biopsy

biopsy

biopsy

biopsy

biopsy

biopsy

biopsy

biopsy

Core or or Core

excision

Trephine Trephine

Trephine Trephine

Trephine Trephine

Trephine Trephine

site site

Skin

Skin

Liver

node node

node

Bone Bone

Right Right

Bone Bone

Bone Bone

Bone Bone

lymph lymph

lymph lymph

Biopsy Biopsy

Cervical Cervical

inguinal inguinal

marrow marrow

marrow marrow

marrow marrow

marrow marrow

RHOA

Positive

Positive

Positive

Positive

Positive

Positive

Positive

Positive

Positive

mutation

M

M

M

M

M

M

76/

81/

77/

82/

82/

72/

Sex

66/F

66/F

64/F

Age/

5

4 4

1

2

1

-

-

-

-

-

C9

C5

C2

C17

C13

C11

C7

C7

C7

C3 C3 Case

up up

, ,

On On

cell cell

-

diagnostic

biopsy. biopsy.

-

lymphoma, lymphoma,

lymphoma, not not lymphoma,

considered

cell cell

-

advised for follow follow for advised

progresses

mutation positivity mutation

cell cell

T

-

cell hyperplasia cell

T

-

AITL is is AITL

evidence

diagnostic

lymphoma

I I

-

biopsy if lymphadenopathy lymphadenopathy if biopsy

RHOA RHOA

-

cell process with plasmacytoid plasmacytoid with cell process

EBV positive polymorphous polymorphous positive EBV

-

persists or or persists

B

Reactive T Reactive

pattern

cell lymphoma, but not not but lymphoma, cell

The component looks like like looks component cell B The

-

Suspicious a of Suspicious

Clonal Clonal

Not diagnostic, not excluding B excluding not diagnostic, Not

Suspicious a of Suspicious

a T a

with a low threshold re for threshold low a with

review with with review

associated LPD and and LPD associated immunodeficiency

small tissue and lack of strong molecular molecular strong of lack and tissue small

A clonal clonal A

differentiation, and not entirely excluding excluding entirely not and differentiation,

there is a relatively small TFH component. component. TFH small relatively is a there

Advised for re for Advised

particularly AITL, but not diagnostic to due diagnostic not but AITL, particularly

lymphoproliferation, lymphoproliferation,

n/a

Weak Weak

Weak Weak

clonal clonal

clonal clonal

Clonal

Clonal

Clonal

Clonal

-

-

re

re

Weak Weak

Clonal

clonal clonal

clonal clonal

Polyclonal Polyclonal

polyclonal polyclonal

Weak clonal Weak

Weak clonal Weak

arrangements

arrangements

Weak multiple multiple Weak

Weak multiple multiple Weak

, ,

. .

cell cell

-

is a a is

cell cell

T

-

cells

+

cells are cells

T cells T

is loss of of is loss

appeared appeared

follicular follicular

+

+

T cells are are cells T

There There

scattered scattered

expression. expression.

ISH negative ISH

staining is staining

EBER

with slightly slightly with

-

center

The The

There There

/PD1

+

PD1 PD1

spill beyond the beyond spill

EBER

population. There population.

and CD8 and

. .

differentiation

+

+

centers

meshworks

-

cells spilling out of the the of out spilling cells

cell marker marker cell

cell cell

B cells cells B

+

and show partial loss of of loss show partial and

-

-

ISH highlights highlights ISH

+

-

FDC

show no loss of pan T pan of loss no show

, ,

T cells T

+

germinal centre localization, localization, centre germinal

ISH negative ISH

pan T pan

meshworks

-

-

-

cells

centers

. . Some

EBER

with a smaller CD4 smaller a with

T cells T

restriction

+

cell markers (CD20, CD79a, CD19), and and CD19), CD79a, (CD20, markers cell

positive positive

and a population CD4 population of a and

-

germinal germinal

population shows CD10, PD1 and possibly possibly and PD1 CD10, shows population

markers and CD10. Germinal Germinal CD10. and markers

The plasma cells display lambda light chain chain light display lambda cells plasma The

+

. .

CD7 expression in expression CD7

+

cell cell

-

are dominantly CD4 dominantly are

lymphoid cells are mixed CD4 mixed are cells lymphoid

B cells across a range of B of range a across cells B

and showed IG kappa light chain restriction. The restriction. chain light kappa IG showed and

+

slightly ragged. ragged. slightly

+

CD30

are also numerous B cells and scattered scattered and cells B numerous also are

meshworks

-

The The

in a predominantly peri predominantly a in

pan T pan

T cells cells T

express PD1 and show no loss of expression pan of expression of loss show no and PD1 express

, with CD4 expression in some large cells. cells. large some in expression CD4 , with

There There

T cells cells T

T cells T

T cells cells T

A mixture of B and T cells in a vaguely nodular intermixed intermixed nodular vaguely a in cells Tand B of mixture A

This is a mostly necrotic sample. In viable areas, a marked marked a areas, viable In sample. necrotic mostly a is This

striking paracortical hyperplasia by small lymphocytes and and lymphocytes small by hyperplasia paracortical striking

diffuse diffuse EBER

+

is no significant CD30, PD1, CD10, BCL6 expression in the T cell cell T the in expression BCL6 CD10, PD1, CD30, no significant is

BCL6 expression. There is FDC expansion and a high number of of number high a expansion and FDC is There expression. BCL6

The nodal architecture is effaced by a diffuse mixed infiltrate of diffuse infiltrate mixed a effaced by is architecture nodal The

CD7. The smaller CD4 smaller The CD7.

markers, and appear negative for CD10 and BCL6. and CD10 for negative appear and markers,

plasma cells. cells. plasma

which are also also are which

cells were EBER were cells

cells. B cells express pan B pan express cells B cells.

follicles and show CD30 expression. EBER expression. CD30 follicles show and

Multiple perivascular infiltrates comprising a mixed population population of mixed a comprising infiltrates perivascular Multiple

histiocytes. histiocytes.

Lymph node architecture appears essentially preserved. preserved. essentially appears architecture node Lymph

helper helper

the interfollicular area by a polymorphous population population lymphoid of polymorphous a by area interfollicular the

negative for CD10 and BCL6 and these large large these and BCL6 and CD10 for negative

proliferation of variably sized blood vessels is noted. is noted. vessels blood sized variably of proliferation

expanded FDC expanded

MUM1, but negative for CD10 and BCL6. A high proportion of the B B the of proportion high A BCL6. and CD10 for negative but MUM1,

markers. markers.

lymphoid follicles appeared to be reactive and showed no apparent apparent no showed and reactive be to follicles appeared lymphoid

distribution. T cells are predominantly CD8 predominantly are cells T distribution.

They are predominantly CD8 predominantly are They

small to medium sized with preserved preserved with sized medium to small

CD8

Partial effacement of the lymph node architecture and expansion expansion and of architecture node lymph the of effacement Partial

There are multiple, variably preserved germinal germinal preserved variably multiple, are There

expansion of TFH cells, with only a few CD10 few a only with cells, expansion TFHof

small, intermediate and large lymphoid cells. There is mixed CD4 and and CD4 mixed is There cells. lymphoid large and intermediate small,

largely restricted to follicular follicular to restricted largely

which express express which

population. There is expansion expansion FDC of is There population.

Many Many

small to medium atypical lymphoid cells admixed with histiocytes and histiocytes with admixed cells lymphoid atypical medium to small

C

C

C

C

C

C

C

Follow up Follow

Preceding

Preceding

Preceding

Preceding

Preceding

Preceding

Core Core

Core Core

Core Core

Core Core

Punch Punch

biopsy

biopsy

biopsy

biopsy

biopsy

Excision

Excision

Skin

node

node

node

node

node

node

lymph lymph

lymph lymph

lymph lymph

Lymph Lymph

Lymph Lymph

Lymph Lymph

Cervical Cervical

Cervical Cervical

Cervical Cervical

Positive

Positive

Positive

Positive

Positive

Positive

Positive

64/M

80/M

80/M

81/M

79/M

67/M

2

1

3

1

-

-

-

-

C43

C30

C19

C38

C38 C37 C37

SUPPLEMENTARY FIGURES

Figure S1. Representative LNA-PNA qPCR plots for detection of RHOA p.Gly17Val.

A) The qPCR is highly specific for RHOA (c.50G>T; p.Gly17Val) mutation, not detecting any other RHOA changes.

B) Examples of qPCR for detection of RHOA (c.50G>T; p.Gly17Val) mutation in the presence or absence of PNA probe which represses the amplification of the wild-type RHOA sequence, thus boosting the detection of the RHOA mutation. As demonstrated by the total probe amplification curve shift in presence of PNA clamp, while the mutant probe amplification curve is enhanced.

C) Examples of qPCR for detection of RHOA (c.50G>T; p.Gly17Val) mutation using crude DNA preparations.

D) Examples of qPCR for detection of RHOA (c.50G>T; p.Gly17Val) mutation for clonal hematopoiesis of indeterminate potential (CHIP) cases with both TET2 and DNMT3A mutations.

Figure S2: Detection of the AITL cells by BaseScope in situ hybridization (ISH) using a specific probe to the unique VDJ junction sequence of the lymphoma cells (TRB-V5 clone), and qPCR of RHOA Gly17Val in case 7.

Left panel: Representative images of BaseScope-ISH;

Right panel: RHOA Gly17Val qPCR plots for various biopsies of case 7.