Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells

Paul F Robbins, Yong-Chen Lu, Mona El-Gamil, Yong F Li, Colin Gross, Jared Gartner, Jimmy C Lin, Jamie K Teer, Paul Cliften, Eric Tycksen, Yardena Samuels, Steven A Rosenberg, Paul F Robbins, Yong-Chen Lu, Mona El-Gamil, Yong F Li, Colin Gross, Jared Gartner, Jimmy C Lin, Jamie K Teer, Paul Cliften, Eric Tycksen, Yardena Samuels, Steven A Rosenberg

Abstract

Substantial regressions of metastatic lesions have been observed in up to 70% of patients with melanoma who received adoptively transferred autologous tumor-infiltrating lymphocytes (TILs) in phase 2 clinical trials. In addition, 40% of patients treated in a recent trial experienced complete regressions of all measurable lesions for at least 5 years following TIL treatment. To evaluate the potential association between the ability of TILs to mediate durable regressions and their ability to recognize potent antigens that presumably include mutated gene products, we developed a new screening approach involving mining whole-exome sequence data to identify mutated proteins expressed in patient tumors. We then synthesized and evaluated candidate mutated T cell epitopes that were identified using a major histocompatibility complex-binding algorithm for recognition by TILs. Using this approach, we identified mutated antigens expressed on autologous tumor cells that were recognized by three bulk TIL lines from three individuals with melanoma that were associated with objective tumor regressions following adoptive transfer. This simplified approach for identifying mutated antigens recognized by T cells avoids the need to generate and laboriously screen cDNA libraries from tumors and may represent a generally applicable method for identifying mutated antigens expressed in a variety of tumor types.

Figures

Figure 1. Response of TIL 2098 to…
Figure 1. Response of TIL 2098 to candidate epitopes identified from autologous tumors
(a) A screening assay was carried out to evaluate the release of IFN-γ from TIL 2098 in an overnight co-culture with peptide-pulsed T2 cells that were pulsed individually with the top 61 candidate HLA-A*0201 binding peptides (Supplementary Table 1) identified from 2098 mel, with the exception of peptides 9,10,14,23,24,31,33 and 37, which overlapped with peptides 1,4,4,25,12,15,16 and 4, respectively (NT). Peptides 39-62 (Supplementary Table 1) stimulated the release of 100 pg/ml or less of IFN-γ from TIL 2098 and are not depicted in this graph. The autologous 2098 mel stimulate the release of 10,000 pg/ml of IFN-γ from 2098 TIL in this assay. (b-d) T2 cells were pulsed with tittered doses of the indicated mutated or wild type (WT) peptides, and IFN-γ release measured in an overnight co-culture with TIL 2098. (e) Stable transfected of COS7 cells (COS-A2) expressing HLA-A*0201 were transiently transfected with the indicated transcripts and evaluated for their ability to stimulate IFN-γ release from TIL 2098 in an overnight co-culture. (f) 293 cells were transiently transfected with a construct encoding HLA-*0201 as well as the indicated transcripts and evaluated for their ability to stimulate IFN-γ release from TIL 2098 in an overnight co-culture.
Figure 2. Response of TIL 2369 to…
Figure 2. Response of TIL 2369 to candidate epitopes identified from autologous tumors
(a) A screening assay was carried out to evaluate the release of IFN-γ from TIL 2369 in an overnight co-culture with peptide-pulsed 293-A1 cells that were pulsed individually with the top 56 candidate HLA-A*0101-binding peptides identified from 2369 mel (Supplementary Table 2), with the exception of peptides 35, 39 and 44, which overlapped with peptides 20, 33 and 43, respectively, and were not tested (NT). Peptides 24-56 (Supplementary Table 2) stimulated the release of less than 100 pg/ml of IFN-γ from TIL 2369 and are not depicted in this graph. The autologous 2369 mel stimulated the release of 20,838 pg/ml of IFN-γ from TIL 2369 in this assay. (b,c) 293-A1 cells were pulsed with the indicated concentrations of peptides in an overnight co-culture with TIL 2098, and IFN-γ release measured. d. 293-A1 cells were transiently transfected with the indicated constructs and evaluated for their ability to stimulate IFN-γ release from TIL 2369 in an overnight co-culture.
Figure 3. Response of TIL 3309 to…
Figure 3. Response of TIL 3309 to candidate epitopes identified from autologous tumors
(a) A screening assay was carried out to evaluate the release of IFN-γ from TIL 3309 in an overnight co-culture with COS-A11 cells that were pulsed individually with the top 46 candidate HLA-A*1101-binding peptides identified from 3309 mel (Supplementary Table 5). Peptides 37-46 (Supplementary Table 5) stimulated the release of less than 100 pg/ml of IFN-γ from TIL 3309 and are not depicted in this graph. The autologous 3309 mel stimulated the release of 4,600 pg/ml of IFN-γ from TIL 3309 in this assay. (b,c) COS7 cells stably transduced with a retroviral vector expressing HLA-A*1101 (COS-A11) were pulsed with the indicated concentrations of peptides in an overnight co-culture with TIL 3309, and IFN-γ release measured. d. COS-A11 cells were transiently transfected with the indicated constructs and evaluated for their ability to stimulate IFN-γ release from TIL 3309 in an overnight co-culture.
Figure 4. IFN-γ ELISPOT responses of TIL…
Figure 4. IFN-γ ELISPOT responses of TIL and PBMC obtained prior to and following autologous TIL transfer
Samples of PBMC prior to adoptive TIL transfer as well as samples of PBMC obtained approximately one month following transfer were evaluated along with samples of infused TIL. A pre-treatment sample was not available for subject 1, and the pre-treatment sample analyzed from subject 2 was obtained approximately one month following an adoptive TIL transfer that was administered in the absence of non-myeloablative chemotherapy one month prior to infusion of the TIL analyzed in (b). TIL 2098 from subject 1 was cultured with HLA-A*0201+ COS-7 cells (a), TIL 2369 from subject 2 was cultured with HLA*0101+ COS7 cells (b), and TIL 3309 from subject 3 were cultured with HLA*1101+ COS-7 cells (c) that were pulsed with candidate nonamer peptides for two hours at 370C. T cells were also cultured with autologous as well as allogeneic tumor cells, and in addition, each of the individual T cell populations were stimulated in parallel cultures with PMA plus ionomycin (PMA/I). The numbers of IFN-γ ELISPOTS per 105 T cells generated in responses to PMA/I were as follows: Subject 1 TIL 2098:7,300, subject 1 post transfer PBMC:4,700, Subject 2 TIL 2369:21,800, Subject 2 pre-transfer PBMC:21,800, Subject 2 post-transfer PBMC:11,000, Subject 3 TIL 3309:7,700, Subject 3 pre-transfer PBMC: 12,500, Subject 3 post-transfer PBMC:26,300. TIL 2098 was plated at 2,000 cells per well, TIL 2369 and 3309 were plated at 1,000 cells per well, and PBMC were plated at 100,000 cells per well, whereas all of the groups stimulated with PMA/I were plated at 1,000 cells per well. The error bars represent the mean + and − the standard error of the mean of three replicate wells per group.

References

    1. Dudley ME, et al. Cancer Regression and Autoimmunity in Patients After Clonal Repopulation with Antitumor Lymphocytes. Science. 2002;298:850–854.
    1. Dudley ME, et al. Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens. J Clin Oncol. 2008;26:5233–5239.
    1. Rosenberg SA, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res. 2011;17:4550–4557.
    1. Nielsen M, et al. NetMHCpan, a method for quantitative predictions of peptide binding to any HLA-A and -B locus protein of known sequence. PLoS One. 2007;2:e796.
    1. Marincola FM, et al. Locus-specific analysis of human leukocyte antigen class I expression in melanoma cell lines. J Immunother Emphasis Tumor Immunol. 1994;16:13–23.
    1. Salter RD, Cresswell P. Impaired assembly and transport of HLA-A and -B antigens in a mutant TxB cell hybrid. EMBO J. 1986;5:943–949.
    1. Amit S, et al. Axin-mediated CKI phosphorylation of beta-catenin at Ser 45: a molecular switch for the Wnt pathway. Genes Dev. 2002;16:1066–1076.
    1. Zhou J, Dudley ME, Rosenberg SA, Robbins PF. Persistence of multiple tumor-specific T-cell clones is associated with complete tumor regression in a melanoma patient receiving adoptive cell transfer therapy. J Immunother. 2005;28:53–62.
    1. Goshima G, Mayer M, Zhang N, Stuurman N, Vale RD. Augmin: a protein complex required for centrosome-independent microtubule generation within the spindle. J Cell Biol. 2008;181:421–429.
    1. Rammensee H, Bachmann J, Emmerich NP, Bachor OA, Stevanovic S. SYFPEITHI: database for MHC ligands and peptide motifs. Immunogenetics. 1999;50:213–219.
    1. Morel S, et al. Processing of some antigens by the standard proteasome but not by the immunoproteasome results in poor presentation by dendritic cells. Immunity. 2000;12:107–117.
    1. Chapiro J, et al. Destructive cleavage of antigenic peptides either by the immunoproteasome or by the standard proteasome results in differential antigen presentation. J Immunol. 2006;176:1053–1061.
    1. Guillaume B, et al. Two abundant proteasome subtypes that uniquely process some antigens presented by HLA class I molecules. Proc Natl Acad Sci U S A. 2010;107:18599–18604.
    1. Kahle JJ, et al. Comparison of an expanded ataxia interactome with patient medical records reveals a relationship between macular degeneration and ataxia. Hum Mol Genet. 20:510–527.
    1. Blazek D, et al. The Cyclin K/Cdk12 complex maintains genomic stability via regulation of expression of DNA damage response genes. Genes Dev. 25:2158–2172.
    1. Kawakami Y, et al. Cloning of the gene coding for a shared human melanoma antigen recognized by autologous T cells infiltrating into tumor. Proc Natl Acad Sci U S A. 1994;91:3515–3519.
    1. van der Bruggen P, et al. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science. 1991;254:1643–1647.
    1. Boel P, et al. BAGE: a new gene encoding an antigen recognized on human melanomas by cytolytic T lymphocytes. Immunity. 1995;2:167–175.
    1. Robbins PF, et al. A mutated beta-catenin gene encodes a melanoma-specific antigen recognized by tumor infiltrating lymphocytes. J Exp Med. 1996;183:1185–1192.
    1. Cox AL, et al. Identification of a peptide recognized by five melanoma-specific human cytotoxic T cell lines. Science. 1994;264:716–719.
    1. Pieper R, et al. Biochemical identification of a mutated human melanoma antigen recognized by CD4(+) T cells. J Exp Med. 1999;189:757–766.
    1. van der Bruggen P, Stroobant V, Vigneron N, Van den Eynde B. Peptide database: T cell-defined tumor antigens. Cancer Immun. 2012
    1. Matsushita H, et al. Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting. Nature. 482:400–404.
    1. Castle JC, et al. Exploiting the mutanome for tumor vaccination. Cancer Res. 2012;72:1081–1091.
    1. Kvistborg P, et al. TIL therapy broadens the tumor-reactive CD8(+) T cell compartment in melanoma patients. Oncoimmunology. 1:409–418.
    1. Jones S, et al. Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science. 2010;330:228–231.
    1. Greenman C, et al. Patterns of somatic mutation in human cancer genomes. Nature. 2007;446:153–158.
    1. Berger MF, et al. Melanoma genome sequencing reveals frequent PREX2 mutations. Nature. 2012;485:502–506.
    1. Topalian SL, Muul LM, Solomon D, Rosenberg SA. Expansion of human tumor infiltrating lymphocytes for use in immunotherapy trials. J.Immunol.Meth. 1987;102:127–141.
    1. Dudley ME, et al. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J Clin Oncol. 2005;23:2346–2357.
    1. Arnold D, et al. Proteasome subunits encoded in the MHC are not generally required for the processing of peptides bound by MHC class I molecules. Nature. 1992;360:171–174.

Source: PubMed

3
Abonnieren