Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Multiomic profiling of T cell lymphoma after therapy with anti-BCMA CAR T cells and GPRC5D-directed bispecific antibody

Abstract

Chimeric antigen receptor (CAR) T cells and bispecific T cell engagers have become integral components in the treatment of relapsed/refractory multiple myeloma. We report a 63-year-old male who received ciltacabtagene autoleucel CAR T cells and the GPRC5D × CD3 bispecific talquetamab for early relapse of his multiple myeloma. Nine months after CAR T therapy, he developed a symptomatic leukemic peripheral T cell lymphoma with cutaneous and intestinal involvement. Longitudinal single-cell RNA and T cell receptor sequencing of peripheral blood and bone marrow revealed two hyperexpanded CAR-carrying T cell clones. These expanded clones exhibited an exhausted effector-memory T cell transcriptional signature, and the neoplasm itself was sensitive to dexamethasone treatment. The immunophenotypic and transcriptional alterations of these abnormal T cells resembled those of T-large granular lymphocytic leukemia. Spatial transcriptomes of skin lesions confirmed the aberrant CAR-expressing T cells. Whole-genome sequencing revealed three distinct integration sites, within the introns of ZGPAT, KPNA4 and polycomb-associated noncoding RNAs. Before and after CAR T whole-genome analyses implicated clonal outgrowth of a TET2-mutated precursor propelled by additional subclone-specific loss of heterozygosity and other secondary mechanisms. This case highlights the evolution of a CAR-carrying peripheral T cell lymphoma following CAR T cell and bispecific T cell engager therapy, offering critical insights into the clonal evolution from a predisposed hematopoietic precursor to a mature neoplasm.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Persistent CAR T cell proliferation with cutaneous and gastrointestinal manifestations after cilta-cel therapy in a patient with MM.
Fig. 2: Spatially resolved single-cell multiomics analyses uncover clonal diversity and transcriptional shifts in CAR+ T cell lymphoma after steroid therapy.
Fig. 3: WGS reveals TET2 loss-of-function mutations as a common variant in the CAR+ T cell proliferations, while both CAR+ T cell clones exhibit distinct integration sites.
Fig. 4: Clonal outgrowth of a TET2-mutated precursor propelled by additional subclone-specific LOH and other secondary mechanisms.

Similar content being viewed by others

Data availability

The single-cell sequencing data supporting the study’s findings have been deposited in the Gene Expression Omnibus under accession codes GSE283144 (scRNA-seq) and GSE283145 (spatial transcriptome). Preprocessed Seurat Objects for the single-cell data are available via Zenodo at https://zenodo.org/records/14251447 (ref. 67). Access requests to the raw WGS data (FASTQ and BAM files) will be handled according to German law. The first author, T.B. (contact: till.braun@uk-koeln.de), will facilitate external data requests with an expected response time frame of 4 weeks. To ensure the reproducibility of the CAR integration site analysis and genomic variant calling, we provide full access to the relevant minimal datasets via the GitHub repository: https://github.com/fraunhofer-izi/Braun_et_al_2024.

Code availability

Processing and analysis code related to this study is deposited in the GitHub repository at https://github.com/fraunhofer-izi/Braun_et_al_2024.

References

  1. Munshi, N. C. et al. Idecabtagene vicleucel in relapsed and refractory multiple myeloma. N. Engl. J. Med. 384, 705–716 (2021).

    Article  CAS  PubMed  Google Scholar 

  2. Rodriguez-Otero, P. et al. Ide-cel or standard regimens in relapsed and refractory multiple myeloma. N. Engl. J. Med. 388, 1002–1014 (2023).

    Article  CAS  PubMed  Google Scholar 

  3. San-Miguel, J. et al. Cilta-cel or standard care in lenalidomide-refractory multiple myeloma. N. Engl. J. Med. 389, 335–347 (2023).

    Article  CAS  PubMed  Google Scholar 

  4. Shah, N., Chari, A., Scott, E., Mezzi, K. & Usmani, S. Z. B-cell maturation antigen (BCMA) in multiple myeloma: rationale for targeting and current therapeutic approaches. Leukemia 34, 985–1005 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Parikh, R. H. & Lonial, S. Chimeric antigen receptor T-cell therapy in multiple myeloma: a comprehensive review of current data and implications for clinical practice. CA Cancer J. Clin. 73, 275–285 (2023).

    Article  PubMed  Google Scholar 

  6. Morris, E. C., Neelapu, S. S., Giavridis, T. & Sadelain, M. Cytokine release syndrome and associated neurotoxicity in cancer immunotherapy. Nat. Rev. Immunol. 22, 85–96 (2022).

    Article  CAS  PubMed  Google Scholar 

  7. Hamilton, M. P. et al. Risk of second tumors and T-cell lymphoma after CAR T-cell therapy. N. Engl. J. Med. 390, 2047–2060 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Elsallab, M. et al. Second primary malignancies after commercial CAR T-cell therapy: analysis of the FDA Adverse Events Reporting System. Blood 143, 2099–2105 (2024).

    Article  CAS  PubMed  Google Scholar 

  9. Kobbe, G. et al. Aggressive lymphoma after CD19 CAR T-cell therapy. N. Engl. J. Med. 391, 1217–1226 (2024).

    Article  CAS  PubMed  Google Scholar 

  10. Ozdemirli, M. et al. Indolent CD4+ CAR T-cell lymphoma after cilta-cel CAR T-cell therapy. N. Engl. J. Med. 390, 2074–2082 (2024).

    Article  CAS  PubMed  Google Scholar 

  11. Ghilardi, G. et al. T cell lymphoma and secondary primary malignancy risk after commercial CAR T cell therapy. Nat. Med. 30, 984–989 (2024).

    Article  CAS  PubMed  Google Scholar 

  12. Levine, B. L. et al. Unanswered questions following reports of secondary malignancies after CAR-T cell therapy. Nat. Med. 30, 338–341 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Chari, A. et al. Talquetamab, a T-cell-redirecting GPRC5D bispecific antibody for multiple myeloma. N. Engl. J. Med. 387, 2232–2244 (2022).

    Article  CAS  PubMed  Google Scholar 

  14. Moignet, A. & Lamy, T. Latest advances in the diagnosis and treatment of large granular lymphocytic leukemia. Am. Soc. Clin. Oncol. Educ. Book 38, 616–625 (2018).

    Article  PubMed  Google Scholar 

  15. Muhowski, E. M. & Rogers, L. M. Dual TCR-expressing T cells in cancer: how single-cell technologies enable new investigation. Immunohorizons 7, 299–306 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Wiestner, A. et al. Differential gene expression in CLL cells from bone marrow and peripheral blood suggests a role of bone marrow stroma in leukemic cell proliferation. Blood 106, 708 (2005).

    Article  Google Scholar 

  17. Sakhinia, E. et al. Comparison of gene-expression profiles in parallel bone marrow and peripheral blood samples in acute myeloid leukaemia by real-time polymerase chain reaction. J. Clin. Pathol. 59, 1059–1065 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Huuhtanen, J. et al. Single-cell characterization of leukemic and non-leukemic immune repertoires in CD8+ T-cell large granular lymphocytic leukemia. Nat. Commun. 13, 1981 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. de Mel, S., Hue, S. S.-S., Jeyasekharan, A. D., Chng, W.-J. & Ng, S.-B. Molecular pathogenic pathways in extranodal NK/T cell lymphoma. J. Hematol. Oncol. 12, 33 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  21. Hoffmann, S. et al. A multi-split mapping algorithm for circular RNA, splicing, trans-splicing and fusion detection. Genome Biol.15, R34 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Liu, L., McBride, K. M. & Reich, N. C. STAT3 nuclear import is independent of tyrosine phosphorylation and mediated by importin-α3. Proc. Natl Acad. Sci. USA 102, 8150–8155 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Huang, Y.-H. et al. Genomic and transcriptomic profiling of peripheral T cell lymphoma reveals distinct molecular and microenvironment subtypes. Cell Rep. Med. 5, 101416 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Cheon, H. et al. Genomic landscape of TCRαβ and TCRγδ T-large granular lymphocyte leukemia. Blood 139, 3058–3072 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Lone, W., Alkhiniji, A., Manikkam Umakanthan, J. & Iqbal, J. Molecular insights into pathogenesis of peripheral T cell lymphoma: a review. Curr. Hematol. Malig. Rep. 13, 318–328 (2018).

    Article  PubMed  Google Scholar 

  26. Antoni, L., Sodha, N., Collins, I. & Garrett, M. D. CHK2 kinase: cancer susceptibility and cancer therapy – two sides of the same coin? Nat. Rev. Cancer 7, 925–936 (2007).

    Article  CAS  PubMed  Google Scholar 

  27. Gan, X. et al. PRR5L degradation promotes mTORC2-mediated PKC-δ phosphorylation and cell migration downstream of Gα12. Nat. Cell Biol. 14, 686–696 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Greenplate, A. et al. Genomic profiling of T-Cell neoplasms reveals frequent JAK1 and JAK3 mutations with clonal evasion from targeted therapies. JCO Precis. Oncol. 2, 1–16 (2018).

    Article  Google Scholar 

  29. Wahnschaffe, L. et al. JAK/STAT-activating genomic alterations are a hallmark of T-PLL. Cancers 11, 1833 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Alaggio, R. et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Lymphoid Neoplasms. Leukemia 36, 1720–1748 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Sandberg, Y. et al. Lack of common TCRA and TCRB clonotypes in CD8(+)/TCRαβ(+) T-cell large granular lymphocyte leukemia: a review on the role of antigenic selection in the immunopathogenesis of CD8(+) T-LGL. Blood Cancer J. 4, e172 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Marchand, T., Lamy, T. & Loughran, T. P. A modern view of LGL leukemia. Blood 144, 1910–1923 (2024).

    Article  CAS  PubMed  Google Scholar 

  33. Rade, M. et al. Single-cell multiomic dissection of response and resistance to chimeric antigen receptor T cells against BCMA in relapsed multiple myeloma. Nat. Cancer 5, 1318–1333 (2024).

    Article  CAS  PubMed  Google Scholar 

  34. Montes de Oca, R. et al. Biomarker correlates of response to ciltacabtagene autoleucel in patients with relapsed or refractory multiple myeloma from CARTITUDE-1, a phase 1b/2 open-label study, at the ~3 year follow-up. Blood 142, 2099 (2023).

    Article  Google Scholar 

  35. Schrader, A. et al. Actionable perturbations of damage responses by TCL1/ATM and epigenetic lesions form the basis of T-PLL. Nat. Commun. 9, 697 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Jain, N. et al. TET2 guards against unchecked BATF3-induced CAR T cell expansion. Nature 615, 315–322 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Fraietta, J. A. et al. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature 558, 307–312 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Carty, S. A. et al. The loss of TET2 promotes CD8+ T cell memory differentiation. J. Immunol. 200, 82–91 (2018).

    Article  CAS  PubMed  Google Scholar 

  39. Bowman, R. L. & Levine, R. L. TET2 in normal and malignant Hematopoiesis. Cold Spring Harb. Perspect. Med. 7, a026518 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Oberbeck, S. et al. Non-canonical effector functions of the T-memory-like T-PLL cell are shaped by cooperative TCL1A and TCR signaling. Blood 136, 2786–2802 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Larson, R. C. & Maus, M. V. Recent advances and discoveries in the mechanisms and functions of CAR T cells. Nat. Rev. Cancer 21, 145–161 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Samur, M. K. et al. High-dose melphalan treatment significantly increases mutational burden at relapse in multiple myeloma. Blood 141, 1724–1736 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Vicenti, I. et al. External quality assessment of HIV-1 DNA quantification assays used in the clinical setting in Italy. Sci. Rep. 12, 3291 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. van Snippenberg, W. et al. Triplex digital PCR assays for the quantification of intact proviral HIV-1 DNA. Methods 201, 41–48 (2022).

    Article  PubMed  Google Scholar 

  45. Schecter, J. M. & Fan, X. BCMA-targeted CAR-T cell therapy for multiple myeloma. Worldwide patent WO2022116086A1 (2022).

  46. Neavin, D. et al. Demuxafy: improvement in droplet assignment by integrating multiple single-cell demultiplexing and doublet detection methods. Genome Biol. 25, 94 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Germain, P.-L., Lun, A., Garcia Meixide, C., Macnair, W. & Robinson, M. D. Doublet identification in single-cell sequencing data using scDblFinder. F1000Res. 10, 979 (2021).

    Article  PubMed  Google Scholar 

  48. Bais, A. S. & Kostka, D. Scds: computational annotation of doublets in single-cell RNA sequencing data. Bioinformatics 36, 1150–1158 (2020).

    Article  CAS  PubMed  Google Scholar 

  49. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587.e29 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Borcherding, N., Bormann, N. L. & Kraus, G. scRepertoire: an R-based toolkit for single-cell immune receptor analysis. F1000Res. 9, 47 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Andreatta, M., Berenstein, A. J. & Carmona, S. J. ScGate: marker-based purification of cell types from heterogeneous single-cell RNA-seq datasets. Bioinformatics 38, 2642–2644 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Andreatta, M. et al. Interpretation of T cell states from single-cell transcriptomics data using reference atlases. Nat. Commun. 12, 2965 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Andreatta, M. & Carmona, S. J. UCell: robust and scalable single-cell gene signature scoring. Comput. Struct. Biotechnol. J. 19, 3796–3798 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Fu, R. et al. Clustifyr: an R package for automated single-cell RNA sequencing cluster classification. F1000Res. 9, 223 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Chu, Y. et al. Pan-cancer T cell atlas links a cellular stress response state to immunotherapy resistance. Nat. Med. 29, 1550–1562 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Larsson, L., Franzén, L., Ståhl, P. L. & Lundeberg, J. Semla: a versatile toolkit for spatially resolved transcriptomics analysis and visualization. Bioinformatics 39, btad626 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Shen, W., Le, S., Li, Y. & Hu, F. SeqKit: a cross-platform and ultrafast toolkit for FASTA/Q file manipulation. PLoS ONE 11, e0163962 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Hanssen, F. et al. Scalable and efficient DNA sequencing analysis on different compute infrastructures aiding variant discovery. NAR Genom. Bioinform. 6, lqae031 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Koboldt, D. C. Best practices for variant calling in clinical sequencing. Genome Med. 12, 91 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Callari, M. et al. Intersect-then-combine approach: improving the performance of somatic variant calling in whole exome sequencing data using multiple aligners and callers. Genome Med. 9, 35 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  62. McLaren, W. et al. The Ensembl Variant Effect Predictor. Genome Biol. 17, 122 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Knaus, B. J. & Grünwald, N. J. Vcfr: a package to manipulate and visualize variant call format data in R. Mol. Ecol. Resour. 17, 44–53 (2017).

    Article  CAS  PubMed  Google Scholar 

  64. Rustad, E. H. et al. Mmsig: a fitting approach to accurately identify somatic mutational signatures in hematological malignancies. Commun. Biol. 4, 424 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Chen, X. et al. Manta: rapid detection of structural variants and indels for germline and cancer sequencing applications. Bioinformatics 32, 1220–1222 (2016).

    Article  CAS  PubMed  Google Scholar 

  66. Rausch, T. et al. DELLY: structural variant discovery by integrated paired-end and split-read analysis. Bioinformatics 28, i333–i339 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Braun, T. et al. Multi-omic single-cell dissection of leukemic T-cell lymphoma following CAR T-cell therapy. Zenodo https://doi.org/10.5281/zenodo.14251446 (2023).

Download references

Acknowledgements

This work was partly supported by the imSAVAR project, which received funding from the Innovative Medicine Initiative 2 Joint Undertaking (JU) under grant agreement no. 853988. The JU receives support from the European Union’s Horizon 2020 research and innovation program and EFPIA and JDRF INTERNATIONAL. Further, it was partly supported by the CERTAINTY project funded by the European Union (grant agreement no. 101136379). Views and opinions expressed are those of the author(s) only and do not necessarily reflect those of the European Union or the Health and Digital Executive Agency. Neither the European Union nor the granting authority can be held responsible for them. T.B. is funded by the Deutsche Krebshilfe through a Mildred Scheel Nachwuchszentrum scholarship (grant no. 70113307) and, together with N.P., received research grants from the Sander Stiftung (grant no. 2023.084.1) and the DFG (seq-costs in projects, grant no. PF1028/1-1). M. Herling is supported by a grant from the German José-Carreras Leukemia Foundation (grant no. DJCLS 01 R_2023) and by the Faculty of Medicine of the University of Leipzig (endowed Professorship). T.B. and M. Herling are both part of the ImmuneT-ME consortium (EPPERMED2024-522). M.M. is supported by a Translational Research Award from the International Myeloma Society, the German Research Foundation SPP µbone, the EU Horizon Program CERTAINTY and the Deutsche Jose Carreras Leukämie Stiftung. Additionally, M.M. has received research funding from Janssen, SpringWorks and Roche/Genentech. K.R. is supported by imSAVAR (Innovative Medicine Initiative 2 Joint Undertaking grant no. 853988), CERTAINTY (European Union, grant agreement no. 101136379), T2Evolve (Innovative Medicine Initiative 2 Joint Undertaking grant no. 945393), SaxoCell (BMBF Clusters4Future, grant no. 03ZU111MB/03ZU111MD), DAAD project grant no. 57616814 (SECAI, School of Embedded Composite AI) and the German José-Carreras Leukemia Foundation (grant no. DJCLS 08 R/2023). We extend our gratitude to C.-A. Voltin (University Hospital of Cologne) for providing PET–CT scan images, G. Allo (University Hospital of Cologne) for providing endoscopy images and C. Hertel (University Hospital of Leipzig) for sample handling. We thank S. Greiser (Experimental Imaging Unit, Fraunhofer IZI) for technical support with the microscopy of the H&E slides for spatial transcriptomics. In addition, many thanks to A. Raap, W. Jahnke and J. Wiegand from the Diagnostics Department of the Fraunhofer IZI for preparing samples and conducting WGS and spatial transcriptomic analyses. Most importantly, we express our deepest thanks to the patient for his invaluable contribution.

Author information

Authors and Affiliations

Authors

Contributions

T.B. J.M., E.H., V.S., U.H., C.S. and T.R. were involved in diagnostic and clinical case management. T.B., M.R., K.R., M.M. and M. Herling were responsible for the experimental design. T.B., H.K., F.K. and D.L. performed the experiments. M.R., M.K., D.F., F.G., N.-N.P. and C.K.K were responsible for data analysis. T.B., N.P., U.P., M. Hallek, U.H., U.K., C.S., K.R., M.M. and M. Herling provided resources and supervision. T.B., M.R., K.R., M.M. and M. Herling prepared the manuscript. All authors revised and approved the manuscript.

Corresponding author

Correspondence to Maximilian Merz.

Ethics declarations

Competing interests

M.M. gave advisory boards and received honoraria and research support from Amgen, BMS, Celgene, Gilead, Janssen, Stemline, Springworks, Sanofi and Takeda. U.H. received consultant and/or speaker fees from Bristol-Myers Squibb, Gilead, Janssen, Miltenyi Biotec and Novartis. C.S. gave advisory boards and received honoraria from Amgen, Abbvie, Bristol-Myers Squibb, Janssen, Novartis, Oncopeptides, Pfizer, Roche, Sanofi, Stemline Menarini and Takeda, and received research support from Janssen and Takeda. U.K. received consultant and/or speaker fees from AstraZeneca, Affimed, Glycostem, GammaDelta, Zelluna, CGT manufacturing: Miltenyi Biotec and Novartis Pharma GmbH, Bristol-Myers Squibb GmbH & Co. M. Herling gave advisory boards and received honoraria from Abbvie, Beigene, Jazz, Janssen, Stemline Menarini and Takeda, and received research support from EDO-Mundipharma, Janpix, Novartis and Roche. The other authors declare no competing interests.

Peer review

Peer review information

Nature Medicine thanks Jake Jackson, Simone Webb and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Saheli Sadanand, in collaboration with the Nature Medicine team.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Cutaneous infiltration of clonal CD30+ T cells.

Immunohistochemical analysis (n=1) of a skin biopsy obtained from the left dorsal region of the patient at nine months post CAT-T therapy. The panels display staining for H&E, CD3, CD5, CD4, Granzyme B, MUM1, and CD30, with the magnification indicated in each image. The biopsy revealed dense lymphocytic infiltrations associated with smaller blood vessels. These infiltrates were positive for CD3, MUM1, Granzyme B, and CD30, predominantly negative for CD4, and exhibited partial loss of CD5.

Extended Data Fig. 2 Quantification of CAR Transgene by Digital PCR of the Lentiviral LTR Sequence.

This figure illustrates the results of digital PCR (ddPCR) analysis for quantification of the CAR transgene. The FAM fluorescence amplitude corresponds to the quantification of the lentiviral LTR sequence, indicating CAR transgene expression. The HEX fluorescence amplitude indicates the RPP30 reference gene as a control for normalization. Red dot populations are positive partitions, blue dot populations are negative partitions, and the dashed line marks the threshold for positivity. Quantification is presented for each condition (copies/µl). Upper panel: Results are displayed for skin biopsy, both in undiluted and serially diluted formats (1:10 and 1:100), and peripheral blood (PB). Lower panel: Results are displayed for duodenal biopsy.

Extended Data Fig. 3 Flow cytometry reveals predominant memory T-cell differentiation.

(a) Flow cytometry analysis of lymphocyte populations in PB. Upper Panel: Lymphocytes were identified based on forward scatter (FS) and side scatter (SS) parameters, and subsequently divided into CD5+ and CD5- populations. Middle Panel: Analysis of the CD5+ population, and Lower Panel: Analysis of the CD5- population. In both the middle and lower panels, naïve and memory T cells were identified by CD45RA and CD45RO expression, respectively. Staining for CCR7 and CD62L allowed further subclassification. (b) Flow cytometry analysis of bone marrow 9 months post-CAR-T infusion, at the time of cutaneous lesion development, and prior to dexamethasone treatment. Upper left: Lymphocytes are identified based on side scatter (SS) and CD45 expression. Lower left: CD5+ BCMA-CAR- T cells. Upper right: CD5+ BCMA-CAR+ T cells. Lower right: CD5- BCMA-CAR+ T cells. Expression profiles of TRBC1 and TRBC2, as well as CD4 and CD8, are shown for each population. The percentage of expression for each marker and the corresponding fluorochrome are indicated in the graph. (c) Sorting strategy for flow cytometry-based cell sorting of CAR+CD5+, CAR+CD5-, and CAR- CD5+ T-cell populations. Singlets were selected based on the SS, and viable T-cells based on the expression of CD3+ and Calcein+. From the single Calcein+CD3+ T-cells, the three respective populations were sorted based on the expression of CAR and CD5.

Extended Data Fig. 4 Duodenal infiltration of clonal T cells.

Immunohistochemical analysis (n=1) of a duodenal biopsy performed at twelve months post CAR-T infusion. The panels display staining for H&E, CD3, CD5, CD4, Granzyme B, MUM1, and CD30, with the magnification indicated in each image. The biopsy revealed again dense lymphocytic infiltrations. These infiltrates were positive for CD3, CD5, and Granzyme B, partially positive for CD4, and negative for MUM1 and CD30.

Extended Data Fig. 5 Dimension reduction for CD8 T cells.

(a) CD8 cells were embedded into a two-dimensional space by the UMAP method. Each dot represents a single cell. (b) Cells are colored by CD45RO/RA expression using ADT (antibody-derived tag) data normalized using a centered log ratio transformation (CLR). Expression values <1 are considered as background noise.

Extended Data Fig. 6 Copy-number profile of chromosome 6 for Clone 1_3.

For each clonotype raw expression values were classified into gains (exprs>1.05) and deletions (exprs<0.95). Panel (a) shows the copy-number profile of chromosome 6 for Clone 1_3. The proportion of cells with gains (red) and deletions (blue) is presented in genomic order. (b) Differential gene expression analysis between Clone 1_3, Clone 2, and apheresis (Aph) for genes located in the region with the most recurrent gain (>75% of cells affected in Clone 1_3).

Extended Data Fig. 7 Enrichment analysis with T-cell specific gene sets.

Differences in enrichment scores for T-cell-specific gene sets after dexamethasone treatment, comparing the samples PB with PB+Dexa, by Clone 1_3 and Clone 2. Average enrichment scores were calculated for each gene set and sample, with significance (FDR < 0.05) assessed using a two-sided Wilcoxon test. (* p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.001).

Extended Data Fig. 8 Mutation frequencies in the different T-cell populations at the time point of lymphoma development.

All nonsynonymous somatic single nucleotide variants and indels that passed quality control (see Methods) were included. The filled colors of the bar plot segments represent genomic locations of retained variants for tumor mutational burden (TMB) analysis. TMB was calculated per hg38 effective genome size (3,049 mb; Hg38 7-way Genome size statistics - genomewiki). We detected a higher TMB in the CAR+CD5- (1.9 Mut/Mb) and the CAR+CD5+ (0.8 Mut/Mb) populations compared to the CAR-CD5+ (0.1 Mut/Mb) population. These results showed that TMB in the neoplastic CAR+ clones were within the range of a recently published (Schrader et al Nat Commun 2018) whole exome sequencing (WES, NimbleGen SeqCap EZ Exome v3: 64,000,000 bp) cohort of 17 T-cell prolymphocytic leukemia (T-PLL) patients (median TMB: 1.1 Mut/Mb; range, 0.2 – 2.3) and 2 T-cell large granular lymphocyte (LGL) patients (median TMB: 1.1 Mut/Mb). WGS TMB was consistent after filtering mutations in coding DNA sequences (CDS) and untranslated regions (UTR) of protein-coding genes divided by genome size of the respective regions (84 Mb) in hg38 build using the GENCODE v46 annotations (CAR+CD5-: 2.0 Mut/Mb, CAR+CD5+: 0.7 Mut/Mb; CAR-CD5+: 0.1 Mut/Mb).

Extended Data Fig. 9 Variant allele frequency in scRNA-seq samples.

Single-cell RNA-Seq data was analyzed for reads supporting the TET2 p.R544* variant. Samples of BM-preDexa, PB-preDexa, and PB-postDexa were merged and split into Clone 1_3, Clone 2 and other T-cells (Other clones). For the analysis of the apheresis sample no filtering for T-cells was performed. The frequency of supporting reads is shown for Clone 1_3, Clone 2, other clones and the apheresis sample. For each sample, the number of high-quality supporting reads / total reads is presented. Notably, 3 out of 109 total reads of the TET2 showed the respective variant in the apheresis sample, indicating CHIP origin of this mutation.

Extended Data Table 1 Overview of Samples Utilized in the Study

Supplementary information

Supplementary Information

Supplementary Figs. 1–18 and Tables 1–8.

Reporting Summary

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Braun, T., Rade, M., Merz, M. et al. Multiomic profiling of T cell lymphoma after therapy with anti-BCMA CAR T cells and GPRC5D-directed bispecific antibody. Nat Med 31, 1145–1153 (2025). https://doi.org/10.1038/s41591-025-03499-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41591-025-03499-9

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer