Mature T-cell lymphomagenesis induced by retroviral insertional activation of Janus kinase 1

Tim Heinrich, Benjamin Rengstl, Alexander Muik, Mina Petkova, Frederike Schmid, Robin Wistinghausen, Kathrin Warner, Giuliano Crispatzu, Martin-Leo Hansmann, Marco Herling, Dorothee von Laer, Sebastian Newrzela, Tim Heinrich, Benjamin Rengstl, Alexander Muik, Mina Petkova, Frederike Schmid, Robin Wistinghausen, Kathrin Warner, Giuliano Crispatzu, Martin-Leo Hansmann, Marco Herling, Dorothee von Laer, Sebastian Newrzela

Abstract

Retroviral vectors (RVs) are powerful tools in clinical gene therapy. However, stable genomic integration of RVs can be oncogenic, as reported in several animal models and in clinical trials. Previously, we observed that T-cell receptor (TCR) polyclonal mature T cells are resistant to transformation after gammaretroviral transfer of (proto-)oncogenes, whereas TCR-oligoclonal T cells were transformable in the same setting. Here, we describe the induction of a mature T-cell lymphoma (MTCL) in TCR-oligoclonal OT-I transgenic T cells, transduced with an enhanced green fluorescent protein (EGFP)-encoding gammaretroviral vector. The tumor cells were of a mature T-cell phenotype and serially transplantable. Integration site analysis revealed a proviral hit in Janus kinase 1 (Jak1), which resulted in Jak1 overexpression and Jak/STAT-pathway activation, particularly via signal transducer and activator of transcription 3 (STAT3). Specific inhibition of Jak1 markedly delayed tumor growth. A systematic meta-analysis of available gene expression data on human mature T-cell lymphomas/leukemias confirmed the relevance of Jak/STAT overexpression in sporadic human T-cell tumorigenesis. To our knowledge, this is the first study to describe RV-associated insertional mutagenesis in mature T cells.

Figures

Figure 1
Figure 1
T-cell lymphomagenesis induced by transduction of an EGFP-encoding gammaretroviral control vector. (a) The gammaretroviral vector used in this study comprised long terminal repeats (LTRs) from myeloproliferative sarcoma virus (MPSV) and a leader region from the murine embryonic stem cell virus (MESV leader 71) including a splice donor and a splice acceptor site. A posttranscriptional regulatory element of the Woodchuck hepatitis virus (WPRE) was located 3′ of the transgene enhanced green fluorescent protein (EGFP). (b) T lymphocytes from OT-I donor mice were isolated and transduced with gammaretroviral particles encoding for EGFP and transplanted into RAG1−/−–recipient mice. One year after transplantation, splenocytes and lymphocytes were reisolated and serially transplanted into secondary RAG1−/−–recipient mice. Four months after serial transplantation, mature T-cell lymphoma emerged. (c) Representative histological sections (HE stained) from lymph node, spleen, kidney, liver, and lung of a diseased animal (magnification 200× scale bar = 100 µm). (d) Cell surface characterization of control vector–induced malignancy. After single cell suspension of tumor samples, cells from massively enlarged spleens were stained for the α chain of the OT-I TCR (Vα2), CD3, CD8, and ICOS (inducible co-stimulator). Staining for CD4 was negative. Representative blots (gated on EGFP-positive cells) from the primary tumor are depicted. Cell surface marking was comparable in all secondary and tertiary recipients. Staining of lymph node samples showed the same results.
Figure 2
Figure 2
Serial transplantation of primary tumor cells reveals outgrowth of a T-cell clone with a retroviral insertion site in Jak1. (a) Tumor cells from the primary mature T-cell tumor (1st generation) were serially transplantable into secondary (2nd generation) and tertiary (3rd generation) recipients and established histological and immunophenotypical identical malignancies. 5 × 106 tumor cells from spleen and lymph node were injected intravenously into secondary and tertiary RAG1−/− recipients. (b) Retroviral integration site (RIS) analysis revealed eight unique proviral insertions in the primary tumor, whereof one RIS was detectable in all animals of the three generations. The RIS was amplified via integration site-specific PCR and verified by sequencing. Primary, secondary, and tertiary tumor samples are depicted by 1, 2, and 3, respectively. Negative control (N), untransduced OT-I cells. M, base pair marker. (c) Using ligation-mediated PCR (LM-PCR) and integration site-specific PCR, we identified the proviral insertion in Jak1, that was common to all animals, which succumbed to mature T-cell lymphoma. The EGFP-encoding provirus integrated between exon 1 and exon 2 of the Jak1 gene on chromosome 4. The start codon of the 25 exons spanning the Jak1 gene is depicted on exon 2. Proviral sequence was orientated in sense with Jak1.
Figure 3
Figure 3
Exclusion of an EGFP/Jak1 fusion product and detection of an aberrant transcript. (a) Detection of EGFP by western blot to exclude a fusion-protein of Jak1 and EGFP. EGFP was solely detectable at a molecular weight (MW) of 27 kDa in all diseased animals. This result was confirmed by PCR on cDNA. Primary, secondary, and tertiary tumor samples are depicted by 1, 2, and 3, respectively. Negative control (N), untransduced OT-I T cells. (b) Insertion site and orientation of EGFP-encoding provirus and schematic overview of different detectable Jak1 transcripts in tumor samples. Retroviral insertion site was located on chromosome 4 in the intron between exons 1 and 2 of Jak1 upstream of the start codon, which is depicted on exon 2. Black boxes represent the three 5′ exons of Jak1 (25 exons in total, whereof 24 are coding). Filled arrowheads schematically show the location of the proviral promoter in the 5′ LTR and 3′ LTR, the splice donor (SD), and the splice acceptor (SA) in the leader region (MESV-L), respectively. The presence of different transcripts was investigated by PCR on cDNA. In all recipients, the Jak1 nascent chain transcript (transcript 1) and the transgene transcript (transcript 3) could be detected. In tumor cells from the second generation, we found an aberrant transcript comprising of proviral LTR, the MESV leader region until the SD site and the 3′ located Jak1 exons (transcript 2). Secondary and tertiary tumor samples are depicted by 2 and 3, respectively. Negative control (N), untransduced OT-I T cells. Only vector-related transcripts are shown. Nascent chain transcript was detectable in all investigated samples including controls. M, base pair marker.
Figure 4
Figure 4
Methylation analysis of CpG islands within the proviral LTR in Jak1. DNA methylation analysis was carried out using sodium bisulfite sequencing to determine methylation status of CpG islands within the long terminal repeat (LTR) of the proviral integration in Jak1. The enhancer is located in the U3-region; promoter is spanning the U3-, R-, and U5-region. Filled boxes resemble methylated CpG islands; open boxes resemble non-methylated CpG islands.
Figure 5
Figure 5
Provirus-induced activation of the Jak/STAT-pathway. (a) Western blot analysis showing highly elevated levels of Jak1 in tumor tissue derived from spleen and lymph node carrying the insertion site in Jak1. Primary, secondary, and tertiary tumor samples are depicted by 1, 2, and 3, respectively. Untransduced OT-I cells isolated from RAG1−/− mice were used as negative control N1; ex vivo obtained EGFP-transduced OT-I T cells w/o RIS in Jak1 as negative control N2. (b) Elevated Jak1 RNA expression determined by real-time quantitative PCR. Measurements were performed in duplicates. Relative quantification was based on normalization on the housekeeping gene 18S rRNA. As negative controls, untransduced OT-I cells (N1) isolated from RAG1−/− mice and ex vivo obtained EGFP-transduced OT-I cells (N2) w/o RIS in Jak1 were used. RNA from the primary mature T-cell lymphoma was not isolated. (c) Western blot analysis showing the selective phosphorylation of STAT3 in tumor tissue obtained from spleen and lymph node carrying the RIS in Jak1. Phosphorylation of STAT5 could not be detected. Primary, secondary, and tertiary tumor samples are depicted by 1, 2, and 3, respectively. Untransduced OT-I T cells served as negative control (N).
Figure 6
Figure 6
Reduced tumor growth kinetics in vivo after specific Jak1 inhibition by INCB018424. Survival of animals transplanted with either EGFP control vector–transformed tumor cells, carrying the genetic lesion in Jak1 (n = 16), or ΔTrkA-induced tumor cells (n = 8) under treatment of Jak1/2 inhibitor INCB018424 or vehicle only. Treated as well as untreated animals transplanted with ΔTrkA-induced tumor cells developed hematopoietic malignancies within 8 days. Tumor development in recipients transplanted with T cells bearing the RIS in Jak1 was significantly delayed by the inhibitor INCB018424 (***P = 0.0001, log-rank).
Figure 7
Figure 7
Gene expression levels of Jak and STAT family members in human primary mature T-cell lymphomas. Array-based mRNA expression levels of the Jak and STAT family genes were retrieved from publically available primary in silico datasets of whole-genome profilings and summarized in a heatmap showing upregulation (red) and downregulation (yellow) across various human MTCL entities and subsets of normal T cells. On the X-axis, the first digit(s) indicate(s) the number of samples per entity per dataset; the second field abbreviates the entity followed by the primary data reference. Unsupervised clustering reveals the association of Jak1 with STAT3 by showing the closest resemblance of expression profiles across MTCL and normal T-cell isolates. The predominantly high expression of Jak1 and to a lesser degree of STAT3 in most MTCL except for some anaplastic large-cell lymphomas and normal T cells, emphasizes the relevance of these molecules in spontaneous mature T-cell tumorigenesis. Another Jak family member highly expressed in the analyzed MTCLs is Tyk2, which shows a closer resemblance with the STAT1 pattern. AB HSTL, α-β hepatosplenic T-cell lymphoma; AITL, Angioimmunoblastic T-cell lymphoma; ALCL-ALK−, Anaplastic large-cell lymphoma ALK negative; ALCL-ALK+, Anaplastic large-cell lymphoma ALK positive; ATLL, Adult T-cell lymphoma; cALCL, primary cutaneous anaplastic large cell lymphoma; CTCL, Cutaneous T-cell lymphoma; EATL, Enteropathy-associated T-cell lymphoma; HSTL, hepatosplenic T-cell lymphoma; PTCL-NOS, Peripheral T-cell lymphoma not otherwise specified; T-cells DR+, T-cells DR−, DR: HLA-DR, human leukocyte antigen receptor DR positive/negative; T-PLL, T-cell prolymphocytic leukemia; T-PLL no inv(14), T-cell prolymphocytic leukemia without inv(14)/t(14;14); T-PLL inv (14), T-cell prolymphocytic leukemia with inv(14)/t(14;14); T-cells CD8A, activated CD8 T cells; T-cells CD8R, resting CD8 T cells; T-cells CD4A, activated CD4 T cells; T-cells CD4R, resting CD4 T cells.

References

    1. Wu X, Li Y, Crise B, Burgess SM. Transcription start regions in the human genome are favored targets for MLV integration. Science. 2003;300:1749–1751.
    1. Laufs S, Nagy KZ, Giordano FA, Hotz-Wagenblatt A, Zeller WJ, Fruehauf S. Insertion of retroviral vectors in NOD/SCID repopulating human peripheral blood progenitor cells occurs preferentially in the vicinity of transcription start regions and in introns. Mol Ther. 2004;10:874–881.
    1. Bushman F, Lewinski M, Ciuffi A, Barr S, Leipzig J, Hannenhalli S, et al. Genome-wide analysis of retroviral DNA integration. Nat Rev Microbiol. 2005;3:848–858.
    1. Staal FJ, Pike-Overzet K, Ng YY, van Dongen JJ. Sola dosis facit venenum. Leukemia in gene therapy trials: a question of vectors, inserts and dosage. Leukemia. 2008;22:1849–1852.
    1. Li Z, Düllmann J, Schiedlmeier B, Schmidt M, von Kalle C, Meyer J, et al. Murine leukemia induced by retroviral gene marking. Science. 2002;296:497.
    1. Hacein-Bey-Abina S, Garrigue A, Wang GP, Soulier J, Lim A, Morillon E, et al. Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J Clin Invest. 2008;118:3132–3142.
    1. Howe SJ, Mansour MR, Schwarzwaelder K, Bartholomae C, Hubank M, Kempski H, et al. Insertional mutagenesis combined with acquired somatic mutations causes leukemogenesis following gene therapy of SCID-X1 patients. J Clin Invest. 2008;118:3143–3150.
    1. Ott MG, Schmidt M, Schwarzwaelder K, Stein S, Siler U, Koehl U, et al. Correction of X-linked chronic granulomatous disease by gene therapy, augmented by insertional activation of MDS1-EVI1, PRDM16 or SETBP1. Nat Med. 2006;12:401–409.
    1. Stein S, Ott MG, Schultze-Strasser S, Jauch A, Burwinkel B, Kinner A, et al. Genomic instability and myelodysplasia with monosomy 7 consequent to EVI1 activation after gene therapy for chronic granulomatous disease. Nat Med. 2010;16:198–204.
    1. Avedillo Díez I, Zychlinski D, Coci EG, Galla M, Modlich U, Dewey RA, et al. Development of novel efficient SIN vectors with improved safety features for Wiskott-Aldrich syndrome stem cell based gene therapy. Mol Pharm. 2011;8:1525–1537.
    1. Bonini C, Grez M, Traversari C, Ciceri F, Marktel S, Ferrari G, et al. Safety of retroviral gene marking with a truncated NGF receptor. Nat Med. 2003;9:367–369.
    1. Scholler J, Brady TL, Binder-Scholl G, Hwang WT, Plesa G, Hege KM, et al. Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells. Sci Transl Med. 2012;4:132ra53.
    1. Recchia A, Bonini C, Magnani Z, Urbinati F, Sartori D, Muraro S, et al. Retroviral vector integration deregulates gene expression but has no consequence on the biology and function of transplanted T cells. Proc Natl Acad Sci USA. 2006;103:1457–1462.
    1. Surh CD, Sprent J. Regulation of mature T cell homeostasis. Semin Immunol. 2005;17:183–191.
    1. Hays EF, Bristol GC, McDougall S, Klotz JL, Kronenberg M. Development of lymphoma in the thymus of AKR mice treated with the lymphomagenic virus SL 3-3. Cancer Res. 1989;49:4225–4230.
    1. Newrzela S, Cornils K, Li Z, Baum C, Brugman MH, Hartmann M, et al. Resistance of mature T cells to oncogene transformation. Blood. 2008;112:2278–2286.
    1. Newrzela S, Cornils K, Heinrich T, Schläger J, Yi JH, Lysenko O, et al. Retroviral insertional mutagenesis can contribute to immortalization of mature T lymphocytes. Mol Med. 2011;17:1223–1232.
    1. Newrzela S, Al-Ghaili N, Heinrich T, Petkova M, Hartmann S, Rengstl B, et al. T-cell receptor diversity prevents T-cell lymphoma development. Leukemia. 2012;26:2499–2507.
    1. Akagi K, Suzuki T, Stephens RM, Jenkins NA, Copeland NG. RTCGD: retroviral tagged cancer gene database. Nucleic Acids Res. 2004;32 Database issue:D523–D527.
    1. Quintás-Cardama A, Vaddi K, Liu P, Manshouri T, Li J, Scherle PA, et al. Preclinical characterization of the selective JAK1/2 inhibitor INCB018424: therapeutic implications for the treatment of myeloproliferative neoplasms. Blood. 2010;115:3109–3117.
    1. Meyer J, Rhein M, Schiedlmeier B, Kustikova O, Rudolph C, Kamino K, et al. Remarkable leukemogenic potency and quality of a constitutively active neurotrophin receptor, deltaTrkA. Leukemia. 2007;21:2171–2180.
    1. Iqbal J, Weisenburger DD, Chowdhury A, Tsai MY, Srivastava G, Greiner TC, International Peripheral T-cell Lymphoma Project et al. Natural killer cell lymphoma shares strikingly similar molecular features with a group of non-hepatosplenic ?d T-cell lymphoma and is highly sensitive to a novel aurora kinase A inhibitor in vitro. Leukemia. 2011;25:348–358.
    1. Iqbal J, Weisenburger DD, Greiner TC, Vose JM, McKeithan T, Kucuk C, International Peripheral T-Cell Lymphoma Project et al. Molecular signatures to improve diagnosis in peripheral T-cell lymphoma and prognostication in angioimmunoblastic T-cell lymphoma. Blood. 2010;115:1026–1036.
    1. Piccaluga PP, Agostinelli C, Califano A, Rossi M, Basso K, Zupo S, et al. Gene expression analysis of peripheral T cell lymphoma, unspecified, reveals distinct profiles and new potential therapeutic targets. J Clin Invest. 2007;117:823–834.
    1. Travert M, Huang Y, de Leval L, Martin-Garcia N, Delfau-Larue MH, Berger F, et al. Molecular features of hepatosplenic T-cell lymphoma unravels potential novel therapeutic targets. Blood. 2012;119:5795–5806.
    1. van Doorn R, van Kester MS, Dijkman R, Vermeer MH, Mulder AA, Szuhai K, et al. Oncogenomic analysis of mycosis fungoides reveals major differences with Sezary syndrome. Blood. 2009;113:127–136.
    1. de Leval L, Rickman DS, Thielen C, Reynies Ad, Huang YL, Delsol G, et al. The gene expression profile of nodal peripheral T-cell lymphoma demonstrates a molecular link between angioimmunoblastic T-cell lymphoma (AITL) and follicular helper T (TFH) cells. Blood. 2007;109:4952–4963.
    1. Dürig J, Bug S, Klein-Hitpass L, Boes T, Jöns T, Martin-Subero JI, et al. Combined single nucleotide polymorphism-based genomic mapping and global gene expression profiling identifies novel chromosomal imbalances, mechanisms and candidate genes important in the pathogenesis of T-cell prolymphocytic leukemia with inv(14)(q11q32). Leukemia. 2007;21:2153–2163.
    1. Shin J, Monti S, Aires DJ, Duvic M, Golub T, Jones DA, et al. Lesional gene expression profiling in cutaneous T-cell lymphoma reveals natural clusters associated with disease outcome. Blood. 2007;110:3015–3027.
    1. Eckerle S, Brune V, Döring C, Tiacci E, Bohle V, Sundström C, et al. Gene expression profiling of isolated tumour cells from anaplastic large cell lymphomas: insights into its cellular origin, pathogenesis and relation to Hodgkin lymphoma. Leukemia. 2009;23:2129–2138.
    1. Lamant L, de Reyniès A, Duplantier MM, Rickman DS, Sabourdy F, Giuriato S, et al. Gene-expression profiling of systemic anaplastic large-cell lymphoma reveals differences based on ALK status and two distinct morphologic ALK+ subtypes. Blood. 2007;109:2156–2164.
    1. Deeks SG, Wagner B, Anton PA, Mitsuyasu RT, Scadden DT, Huang C, et al. A phase II randomized study of HIV-specific T-cell gene therapy in subjects with undetectable plasma viremia on combination antiretroviral therapy. Mol Ther. 2002;5:788–797.
    1. Mitsuyasu RT, Anton PA, Deeks SG, Scadden DT, Connick E, Downs MT, et al. Prolonged survival and tissue trafficking following adoptive transfer of CD4zeta gene-modified autologous CD4(+) and CD8(+) T cells in human immunodeficiency virus-infected subjects. Blood. 2000;96:785–793.
    1. Walker RE, Bechtel CM, Natarajan V, Baseler M, Hege KM, Metcalf JA, et al. Long-term in vivo survival of receptor-modified syngeneic T cells in patients with human immunodeficiency virus infection. Blood. 2000;96:467–474.
    1. Migone TS, Lin JX, Cereseto A, Mulloy JC, O'Shea JJ, Franchini G, et al. Constitutively activated Jak-STAT pathway in T cells transformed with HTLV-I. Science. 1995;269:79–81.
    1. Yu CL, Jove R, Burakoff SJ. Constitutive activation of the Janus kinase-STAT pathway in T lymphoma overexpressing the Lck protein tyrosine kinase. J Immunol. 1997;159:5206–5210.
    1. Flex E, Petrangeli V, Stella L, Chiaretti S, Hornakova T, Knoops L, et al. Somatically acquired JAK1 mutations in adult acute lymphoblastic leukemia. J Exp Med. 2008;205:751–758.
    1. Zhang Q, Raghunath PN, Xue L, Majewski M, Carpentieri DF, Odum N, et al. Multilevel dysregulation of STAT3 activation in anaplastic lymphoma kinase-positive T/null-cell lymphoma. J Immunol. 2002;168:466–474.
    1. Khoury JD, Medeiros LJ, Rassidakis GZ, Yared MA, Tsioli P, Leventaki V, et al. Differential expression and clinical significance of tyrosine-phosphorylated STAT3 in ALK+ and ALK- anaplastic large cell lymphoma. Clin Cancer Res. 2003;9 10 Pt 1:3692–3699.
    1. Sommer VH, Clemmensen OJ, Nielsen O, Wasik M, Lovato P, Brender C, et al. In vivo activation of STAT3 in cutaneous T-cell lymphoma. Evidence for an antiapoptotic function of STAT3. Leukemia. 2004;18:1288–1295.
    1. Schambach A, Wodrich H, Hildinger M, Bohne J, Kräusslich HG, Baum C. Context dependence of different modules for posttranscriptional enhancement of gene expression from retroviral vectors. Mol Ther. 2000;2:435–445.
    1. Newrzela S, Gunda B, von Laer D. T cell culture for gammaretroviral transfer. Methods Mol Biol. 2009;506:71–82.
    1. Li LC, Dahiya R. MethPrimer: designing primers for methylation PCRs. Bioinformatics. 2002;18:1427–1431.

Source: PubMed

3
Abonnieren