CP: stem cell research; cancer immunotherapy; induced pluripotent stem cells; major histocompatibility complex class I; mass spectrometry; tumor-specific antigens; Histocompatibility Antigens Class I; Peptides; Animals; Histocompatibility Antigens Class I/metabolism; Humans; Mice; Peptides/metabolism; Induced Pluripotent Stem Cells; Neoplasms/metabolism; Pluripotent Stem Cells/metabolism; Neoplasms; Pluripotent Stem Cells; Biochemistry, Genetics and Molecular Biology (all); General Biochemistry, Genetics and Molecular Biology
Abstract :
[en] Previous reports showed that mouse vaccination with pluripotent stem cells (PSCs) induces durable anti-tumor immune responses via T cell recognition of some elusive oncofetal epitopes. We characterize the MHC I-associated peptide (MAP) repertoire of human induced PSCs (iPSCs) using proteogenomics. Our analyses reveal a set of 46 pluripotency-associated MAPs (paMAPs) absent from the transcriptome of normal tissues and adult stem cells but expressed in PSCs and multiple adult cancers. These paMAPs derive from coding and allegedly non-coding (48%) transcripts involved in pluripotency maintenance, and their expression in The Cancer Genome Atlas samples correlates with source gene hypomethylation and genomic aberrations common across cancer types. We find that several of these paMAPs were immunogenic. However, paMAP expression in tumors coincides with activation of pathways instrumental in immune evasion (WNT, TGF-β, and CDK4/6). We propose that currently available inhibitors of these pathways could synergize with immune targeting of paMAPs for the treatment of poorly differentiated cancers.
Disciplines :
Biochemistry, biophysics & molecular biology
Author, co-author :
Apavaloaei, Anca; Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada, Department of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
Hesnard, Leslie; Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada
Hardy, Marie-Pierre; Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada
Benabdallah, Basma; CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada
Ehx, Grégory ; Université de Liège - ULiège > Département des sciences cliniques ; Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada
Thériault, Catherine; Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada
Laverdure, Jean-Philippe; Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada
Durette, Chantal; Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada
Lanoix, Joël; Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada
Courcelles, Mathieu; Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada
Noronha, Nandita; Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada, Department of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
Chauhan, Kapil Dev; Faculty of Health Sciences, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
Lemieux, Sébastien; Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada, Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
Beauséjour, Christian; CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada, Department of Pharmacology and Physiology, University of Montreal, Montreal, QC H3T 1J4, Canada
Bhatia, Mick; Faculty of Health Sciences, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
Thibault, Pierre; Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada, Department of Chemistry, University of Montreal, Montreal, QC H3T 1J4, Canada. Electronic address: pierre.thibault@umontreal.ca
Perreault, Claude ; Institute for Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC H3T 1J4, Canada, Department of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada. Electronic address: claude.perreault@umontreal.ca
We wish to thank Qingchuan Zhao, Jean-David Larouche, Assya Trofimov, Maria Virginia Ruiz Cuevas, Eralda Kina, and Krystel Vincent for valuable discussions and suggestions and help with preliminary exploratory analyses. We are grateful to Raphaëlle Lambert, Jennifer Huber, Sarah Boissel, and the IRIC genomics core facility staff for technical assistance with RNA-seq and Patrick Gendron from the IRIC bioinformatics platform for help with RNA-seq and TCGA data analyses. We also thank Dr. Hannah Carter (UC San Diego) for providing the HLA typing information for TCGA patients and the NIH Tetramer Core Facility for providing the tetramers used in this study. Finally, we thank the TCGA and the GTEx Consortium for access to data that enabled this study. This study was supported by grants from the Canadian Cancer Society ( 705604 ) (to C.P. and P.T.), the Canadian Institutes of Health Research ( FDN 148400 ) (to C.P.), The Oncopole (EMC 2 Grant) (to C.P. and P.T.), and the Fonds Vaccins Thérapeutiques Contre le Cancer (to C.P.). A.A. is supported by doctoral studentships from the IRIC , The Cole Foundation , and the Fonds de Recherche du Québec - Santé . For the Bhatia program, this work was supported by the Canada Chair Program (Tier 1) for Human Stem Cell Biology and DeGroote Chair in Stem Cell Biology to M.B. and Canadian Institutes of Health Research (CIHR) foundation grant FRN 159925 to M.B. approved by the Stem Cell Oversight Committee in Canada.We wish to thank Qingchuan Zhao, Jean-David Larouche, Assya Trofimov, Maria Virginia Ruiz Cuevas, Eralda Kina, and Krystel Vincent for valuable discussions and suggestions and help with preliminary exploratory analyses. We are grateful to Raphaëlle Lambert, Jennifer Huber, Sarah Boissel, and the IRIC genomics core facility staff for technical assistance with RNA-seq and Patrick Gendron from the IRIC bioinformatics platform for help with RNA-seq and TCGA data analyses. We also thank Dr. Hannah Carter (UC San Diego) for providing the HLA typing information for TCGA patients and the NIH Tetramer Core Facility for providing the tetramers used in this study. Finally, we thank the TCGA and the GTEx Consortium for access to data that enabled this study. This study was supported by grants from the Canadian Cancer Society (705604) (to C.P. and P.T.), the Canadian Institutes of Health Research (FDN 148400) (to C.P.), The Oncopole (EMC2 Grant) (to C.P. and P.T.), and the Fonds Vaccins Thérapeutiques Contre le Cancer (to C.P.). A.A. is supported by doctoral studentships from the IRIC, The Cole Foundation, and the Fonds de Recherche du Québec - Santé. For the Bhatia program, this work was supported by the Canada Chair Program (Tier 1) for Human Stem Cell Biology and DeGroote Chair in Stem Cell Biology to M.B. and Canadian Institutes of Health Research (CIHR) foundation grant FRN 159925 to M.B. approved by the Stem Cell Oversight Committee in Canada. A.A. M.-P.H. and C.P. designed the study. A.A. performed the cell culture experiments, cytotoxicity assays, main bioinformatic analyses, and data interpretation and wrote the first manuscript draft. C.D. and J.L. performed mass spectrometry experiments. L.H. and C.T. performed in vitro immunogenicity assays. B.B. performed trilineage differentiation assays. M.-P.H. and D.K.C. assisted with preliminary iPSC experiments. N.N. provided assistance with AML cell culture. M.-P.H. G.E. J.-P.L. and M.C. contributed to bioinformatic analyses. M.-P.H. L.H. G.E. B.B. N.N. S.L. C.B. M.B. P.T. and C.P. contributed to the analysis and interpretation of data. All authors edited and approved the final manuscript. A.A. M.-P.H. P.T. and C.P. are named inventors on a patent application filed by Université de Montréal and covering antigens described in this article.
Agudo, J., Park, E.S., Rose, S.A., Alibo, E., Sweeney, R., Dhainaut, M., Kobayashi, K.S., Sachidanandam, R., Baccarini, A., Merad, M., Brown, B.D., Quiescent tissue stem cells evade immune surveillance. Immunity 48 (2018), 271–285.e5, 10.1016/j.immuni.2018.02.001.
Åkesson, E., Wolmer-Solberg, N., Cederarv, M., Falci, S., Odeberg, J., Human neural stem cells and astrocytes, but not neurons, suppress an allogeneic lymphocyte response. Stem Cell Res. 2 (2009), 56–67, 10.1016/j.scr.2008.06.002.
Álvarez-Fernández, M., Malumbres, M., Mechanisms of sensitivity and resistance to CDK4/6 inhibition. Cancer Cell 37 (2020), 514–529, 10.1016/j.ccell.2020.03.010.
Anderson, K.G., Stromnes, I.M., Greenberg, P.D., Obstacles posed by the tumor microenvironment to T cell activity: a case for synergistic therapies. Cancer Cell 31 (2017), 311–325, 10.1016/j.ccell.2017.02.008.
Apavaloaei, A., Hardy, M.P., Thibault, P., Perreault, C., The origin and immune recognition of tumor-specific antigens. Cancers, 12, 2020, 2607, 10.3390/cancers12092607.
Aran, D., Sirota, M., Butte, A.J., Systematic pan-cancer analysis of tumour purity. Nat. Commun. 6 (2015), 8971–9012, 10.1038/ncomms9971.
Aran, D., Camarda, R., Odegaard, J., Paik, H., Oskotsky, B., Krings, G., Goga, A., Sirota, M., Butte, A.J., Comprehensive analysis of normal adjacent to tumor transcriptomes. Nat. Commun. 8 (2017), 1077–1113, 10.1038/s41467-017-01027-z.
Aran, D., Hu, Z., Butte, A.J., xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biol. 18 (2017), 220–314, 10.1186/s13059-017-1349-1.
Babarinde, I.A., Ma, G., Li, Y., Deng, B., Luo, Z., Liu, H., Abdul, M.M., Ward, C., Chen, M., Fu, X., et al. Transposable element-gene splicing modulates the transcriptional landscape of human pluripotent stem cells. Preprint at bioRxiv, 2020, 10.1101/2020.07.26.220608.
Bagati, A., Kumar, S., Jiang, P., Pyrdol, J., Zou, A.E., Godicelj, A., Mathewson, N.D., Cartwright, A.N.R., Cejas, P., Brown, M., et al. Integrin αvβ6–TGFβ–SOX4 pathway drives immune evasion in triple-negative breast cancer. Cancer Cell 39 (2021), 54–67.e9, 10.1016/j.ccell.2020.12.001.
Barbie, D.A., Tamayo, P., Boehm, J.S., Kim, S.Y., Moody, S.E., Dunn, I.F., Schinzel, A.C., Sandy, P., Meylan, E., Scholl, C., et al. Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature 462 (2009), 108–112, 10.1038/nature08460.
Bassani-Sternberg, M., Pletscher-Frankild, S., Jensen, L.J., Mann, M., Mass spectrometry of human leukocyte antigen class I peptidomes reveals strong effects of protein abundance and turnover on antigen presentation. Mol. Cell. Proteomics 14 (2015), 658–673, 10.1074/mcp.M114.042812.
Batlle, E., Clevers, H., Cancer stem cells revisited. Nat. Med. 23 (2017), 1124–1134, 10.1038/nm.4409.
Ben-Porath, I., Thomson, M.W., Carey, V.J., Ge, R., Bell, G.W., Regev, A., Weinberg, R.A., An embryonic stem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nat. Genet. 40 (2008), 499–507, 10.1038/ng.127.
Benhammadi, M., Mathé, J., Dumont-Lagacé, M., Kobayashi, K.S., Gaboury, L., Brochu, S., Perreault, C., IFN-Λ enhances constitutive expression of MHC class I molecules on thymic epithelial cells. J. Immunol. 205 (2020), 1268–1280, 10.4049/jimmunol.2000225.
Bolger, A.M., Lohse, M., Usadel, B., Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30 (2014), 2114–2120, 10.1093/bioinformatics/btu170.
Bouwmeester, R., Gabriels, R., Hulstaert, N., Martens, L., Degroeve, S., DeepLC can predict retention times for peptides that carry as-yet unseen modifications. Nat. Methods 18 (2021), 1363–1369, 10.1038/s41592-021-01301-5.
Brewer, B.G., Mitchell, R.A., Harandi, A., Eaton, J.W., Embryonic vaccines against cancer: an early history. Exp. Mol. Pathol. 86 (2009), 192–197, 10.1016/j.yexmp.2008.12.002.
Caron, E., Vincent, K., Fortier, M.-H., Laverdure, J.-P., Bramoullé, A., Hardy, M.-P., Voisin, G., Roux, P.P., Lemieux, S., Thibault, P., Perreault, C., The MHC I immunopeptidome conveys to the cell surface an integrative view of cellular regulation. Mol. Syst. Biol. 7 (2011), 533–615, 10.1038/msb.2011.68.
Castro, A., Ozturk, K., Pyke, R.M., Xian, S., Zanetti, M., Carter, H., Elevated neoantigen levels in tumors with somatic mutations in the HLA-A, HLA-B, HLA-C and B2M genes. BMC Med. Genom. 12 (2019), 107–113, 10.1186/s12920-019-0544-1.
Chia, N.Y., Chan, Y.S., Feng, B., Lu, X., Orlov, Y.L., Moreau, D., Kumar, P., Yang, L., Jiang, J., Lau, M.S., et al. A genome-wide RNAi screen reveals determinants of human embryonic stem cell identity. Nature 468 (2010), 316–320, 10.1038/nature09531.
Chong, C., Müller, M., Pak, H., Harnett, D., Huber, F., Grun, D., Leleu, M., Auger, A., Arnaud, M., Stevenson, B.J., et al. Integrated proteogenomic deep sequencing and analytics accurately identify non-canonical peptides in tumor immunopeptidomes. Nat. Commun., 11, 2020, 1293, 10.1038/s41467-020-14968-9.
Churko, J.M., Lee, J., Ameen, M., Gu, M., Venkatasubramanian, M., Diecke, S., Sallam, K., Im, H., Wang, G., Gold, J.D., et al. Transcriptomic and epigenomic differences in human induced pluripotent stem cells generated from six reprogramming methods. Nat. Biomed. Eng. 1 (2017), 826–837, 10.1038/s41551-017-0141-6.
Colaprico, A., Silva, T.C., Olsen, C., Garofano, L., Cava, C., Garolini, D., Sabedot, T.S., Malta, T.M., Pagnotta, S.M., Castiglioni, I., et al. TCGAbiolinks: an R/Bioconductor package for integrative analysis of TCGA data. Nucleic Acids Res., 44, 2016, e71, 10.1093/nar/gkv1507.
Coulie, P.G., Van Den Eynde, B.J., Van Der Bruggen, P., Boon, T., Tumour antigens recognized by T lymphocytes: at the core of cancer immunotherapy. Nat. Rev. Cancer 14 (2014), 135–146, 10.1038/nrc3670.
Courcelles, M., Durette, C., Daouda, T., Laverdure, J.-P., Vincent, K., Lemieux, S., Perreault, C., Thibault, P., MAPDP: a cloud-based computational platform for immunopeptidomics analyses. J. Proteome Res. 19 (2020), 1873–1881, 10.1021/acs.jproteome.9b00859.
Dai, Z., Ramesh, V., Locasale, J.W., The evolving metabolic landscape of chromatin biology and epigenetics. Nat. Rev. Genet. 21 (2020), 737–753, 10.1038/s41576-020-0270-8.
Danilova, L., Anagnostou, V., Caushi, J.X., Sidhom, J.W., Guo, H., Chan, H.Y., Suri, P., Tam, A., Zhang, J., Asmar, M.E., et al. The mutation-associated neoantigen functional expansion of specific T cells (MANAFEST) assay: a sensitive platform for monitoring antitumor immunity. Cancer Immunol. Res. 6 (2018), 888–899, 10.1158/2326-6066.CIR-18-0129.
Daouda, T., Perreault, C., Lemieux, S., pyGeno: a Python package for precision medicine and proteogenomics. F1000Res., 5, 2016, 381, 10.12688/F1000RESEARCH.8251.2.
Dardenne, E., Beltran, H., Benelli, M., Gayvert, K., Berger, A., Puca, L., Cyrta, J., Sboner, A., Noorzad, Z., MacDonald, T., et al. N-myc induces an EZH2-mediated transcriptional program driving neuroendocrine prostate cancer. Cancer Cell 30 (2016), 563–577, 10.1016/j.ccell.2016.09.005.
Das, B., Pal, B., Bhuyan, R., Li, H., Sarma, A., Gayan, S., Talukdar, J., Sandhya, S., Bhuyan, S., Gogoi, G., et al. MYC regulates the HIF2a stemness pathway via nanog and Sox2 to maintain self-renewal in cancer stem cells versus non-stem cancer cells. Cancer Res. 79 (2019), 4015–4025, 10.1158/0008-5472.CAN-18-2847.
Duffour, M.T., Chaux, P., Lurquin, C., Cornelis, G., Boon, T., Van Der Bruggen, P., A MAGE-A4 peptide presented by HLA-A2 is recognized by cytolytic T lymphocytes. Eur. J. Immunol. 29 (1999), 3329–3337.
Ehx, G., Perreault, C., Discovery and characterization of actionable tumor antigens. Genome Med. 11 (2019), 29–32, 10.1186/s13073-019-0642-x.
Ellrott, K., Bailey, M.H., Saksena, G., Covington, K.R., Kandoth, C., Stewart, C., Hess, J., Ma, S., Chiotti, K.E., McLellan, M., et al. Scalable open science approach for mutation calling of tumor exomes using multiple genomic pipelines. Cell Syst. 6 (2018), 271–281.e7, 10.1016/j.cels.2018.03.002.
Erhard, F., Dölken, L., Schilling, B., Schlosser, A., Identification of the cryptic HLA-I immunopeptidome. Cancer Immunol. Res. 8 (2020), 1018–1026, 10.1158/2326-6066.CIR-19-0886.
Fagnocchi, L., Zippo, A., Multiple roles of MYC in integrating regulatory networks of pluripotent stem cells. Front. Cell Dev. Biol. 5 (2017), 7–19, 10.3389/fcell.2017.00007.
Fort, A., Hashimoto, K., Yamada, D., Salimullah, M., Keya, C.A., Saxena, A., Bonetti, A., Voineagu, I., Bertin, N., Kratz, A., et al. Deep transcriptome profiling of mammalian stem cells supports a regulatory role for retrotransposons in pluripotency maintenance. Nat. Genet. 46 (2014), 558–566, 10.1038/ng.2965.
Friedli, M., Turelli, P., Kapopoulou, A., Rauwel, B., Castro-Díaz, N., Rowe, H.M., Ecco, G., Unzu, C., Planet, E., Lombardo, A., et al. Loss of transcriptional control over endogenous retroelements during reprogramming to pluripotency. Genome Res. 24 (2014), 1251–1259, 10.1101/gr.172809.114.
Goldman, M.J., Craft, B., Hastie, M., Repečka, K., McDade, F., Kamath, A., Banerjee, A., Luo, Y., Rogers, D., Brooks, A.N., et al. Visualizing and interpreting cancer genomics data via the Xena platform. Nat. Biotechnol. 38 (2020), 675–678, 10.1038/s41587-020-0546-8.
Granados, D.P., Sriranganadane, D., Daouda, T., Zieger, A., Laumont, C.M., Caron-Lizotte, O., Boucher, G., Hardy, M.-P., Gendron, P., Côté, C., et al. Impact of genomic polymorphisms on the repertoire of human MHC class I-associated peptides. Nat. Commun., 5, 2014, 3600, 10.1038/ncomms4600.
Haen, S.P., Löffler, M.W., Rammensee, H.G., Brossart, P., Towards new horizons: characterization, classification and implications of the tumour antigenic repertoire. Nat. Rev. Clin. Oncol. 17 (2020), 595–610, 10.1038/s41571-020-0387-x.
Hänzelmann, S., Castelo, R., Guinney, J., GSVA: gene set variation analysis for microarray and RNA-Seq data. BMC Bioinf., 14, 2013, 7, 10.1186/1471-2105-14-7.
Vander Heiden, M.G., Cantley, L.C., Thompson, C.B., Understanding the warburg effect: the metabolic Requirements of cell proliferation. Science 324 (2009), 1029–1033, 10.1126/science.1160809.
Hernandez, C., Wang, Z., Ramazanov, B., Tang, Y., Mehta, S., Dambrot, C., Lee, Y.W., Tessema, K., Kumar, I., Astudillo, M., et al. Dppa2/4 facilitate epigenetic Remodeling during reprogramming to pluripotency. Cell Stem Cell 23 (2018), 396–411.e8, 10.1016/j.stem.2018.08.001.
Hinze, L., Labrosse, R., Degar, J., Han, T., Schatoff, E.M., Schreek, S., Karim, S., McGuckin, C., Sacher, J.R., Wagner, F., et al. Exploiting the therapeutic interaction of WNT pathway activation and asparaginase for colorectal cancer therapy. Cancer Discov. 10 (2020), 1690–1705, 10.1158/2159-8290.CD-19-1472.
Hong, S.-H., Lee, J.-H., Lee, J.B., Ji, J., Bhatia, M., ID1 and ID3 represent conserved negative regulators of human embryonic and induced pluripotent stem cell hematopoiesis. J. Cell Sci. 124 (2011), 1445–1452, 10.1242/jcs.077511.
Huang, C.-Y., Chung, C.-L., Hu, T.-H., Chen, J.-J., Liu, P.-F., Chen, C.-L., Recent progress in TGF-β inhibitors for cancer therapy. Biomed. Pharmacother., 134, 2021, 111046, 10.1016/j.biopha.2020.111046.
Huang, L.-Q., Brasseur, F., Serrano, A., De Plaen, E., Bruggen, P. van der, Boon, T., Pel, A.V., Cytolytic T lymphocytes recognize an antigen encoded by MAGE-A10 on a human melanoma. J. Immunol. 162 (1991), 6849–6854, 10.1126/science.1840703.
Humeau, J., Sauvat, A., Cerrato, G., Xie, W., Loos, F., Iannantuoni, F., Bezu, L., Lévesque, S., Paillet, J., Pol, J., et al. Inhibition of transcription by dactinomycin reveals a new characteristic of immunogenic cell stress. EMBO Mol. Med., 12, 2020, e11622, 10.15252/emmm.201911622.
Ishak, C.A., De Carvalho, D.D., Reactivation of endogenous retroelements in cancer development and therapy. Annu. Rev. Cell Biol. 4 (2020), 159–176, 10.1146/annurev-cancerbio-030419-033525.
Izadyar, F., Wong, J., Maki, C., Pacchiarotti, J., Ramos, T., Howerton, K., Yuen, C., Greilach, S., Zhao, H.H., Chow, M., et al. Identification and characterization of repopulating spermatogonial stem cells from the adult human testis. Hum. Reprod. 26 (2011), 1296–1306, 10.1093/humrep/der026.
Janku, F., Yap, T.A., Meric-Bernstam, F., Targeting the PI3K pathway in cancer: are we making headway?. Nat. Rev. Clin. Oncol. 15 (2018), 273–291, 10.1038/nrclinonc.2018.28.
Jerby-Arnon, L., Shah, P., Cuoco, M.S., Rodman, C., Su, M.J., Melms, J.C., Leeson, R., Kanodia, A., Mei, S., Lin, J.R., et al. A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell 175 (2018), 984–997.e24, 10.1016/j.cell.2018.09.006.
Jurtz, V., Paul, S., Andreatta, M., Marcatili, P., Peters, B., Nielsen, M., NetMHCpan-4.0: improved peptide–MHC class I interaction predictions integrating eluted ligand and peptide binding affinity data. J. Immunol. 199 (2017), 3360–3368, 10.4049/jimmunol.1700893.
Kazantseva, J., Sadam, H., Neuman, T., Palm, K., Targeted alternative splicing of TAF4: a new strategy for cell reprogramming. Sci. Rep. 6 (2016), 30852–30911, 10.1038/srep30852.
Kelley, D., Rinn, J., Transposable elements reveal a stem cell-specific class of long noncoding RNAs. Genome Biol., 13, 2012, R107, 10.1186/gb-2012-13-11-r107.
Kim, J.Y., Ohn, J., Yoon, J.-S., Kang, B.M., Park, M., Kim, S., Lee, W., Hwang, S., Kim, J.-I., Kim, K.H., Kwon, O., Priming mobilization of hair follicle stem cells triggers permanent loss of regeneration after alkylating chemotherapy. Nat. Commun., 10, 2019, 3694, 10.1038/s41467-019-11665-0.
Klawitter, S., Fuchs, N.V., Upton, K.R., Muñoz-Lopez, M., Shukla, R., Wang, J., Garcia-Cañadas, M., Lopez-Ruiz, C., Gerhardt, D.J., Sebe, A., et al. Reprogramming triggers endogenous L1 and Alu retrotransposition in human induced pluripotent stem cells. Nat. Commun., 7, 2016, 10286, 10.1038/ncomms10286.
Kooreman, N.G., Kim, Y., de Almeida, P.E., Termglinchan, V., Diecke, S., Shao, N.Y., Wei, T.T., Yi, H., Dey, D., Nelakanti, R., et al. Autologous iPSC-based vaccines elicit anti-tumor responses in vivo. Cell Stem Cell 22 (2018), 501–513.e7, 10.1016/j.stem.2018.01.016.
Krokhin, O.V., Sequence-specific retention calculator. Algorithm for peptide retention prediction in ion-pair RP-HPLC: application to 300- and 100-A pore size C18 sorbents. Anal. Chem. 78 (2006), 7785–7795, 10.1021/ac060777w.
Lamoliatte, F., McManus, F.P., Maarifi, G., Chelbi-Alix, M.K., Thibault, P., Uncovering the SUMOylation and ubiquitylation crosstalk in human cells using sequential peptide immunopurification. Nat. Commun., 8, 2017, 14109, 10.1038/ncomms14109.
Larouche, J.D., Trofimov, A., Hesnard, L., Ehx, G., Zhao, Q., Vincent, K., Durette, C., Gendron, P., Laverdure, J.P., Bonneil, É., et al. Widespread and tissue-specific expression of endogenous retroelements in human somatic tissues. Genome Med., 12, 2020, 40, 10.1186/s13073-020-00740-7.
Laumont, C.M., Vincent, K., Hesnard, L., Audemard, É., Bonneil, É., Laverdure, J.-P., Gendron, P., Courcelles, M., Hardy, M.-P., Côté, C., et al. Noncoding regions are the main source of targetable tumor-specific antigens. Sci. Transl. Med., 10, 2018, eaau5516, 10.1126/scitranslmed.aau5516.
Li, L., Baroja, M.L., Majumdar, A., Chadwick, K., Rouleau, A., Gallacher, L., Ferber, I., Lebkowski, J., Martin, T., Madrenas, J., Bhatia, M., Human embryonic stem cells possess immune-privileged properties. Stem Cell. 22 (2004), 448–456, 10.1634/stemcells.22-4-448.
Liberzon, A., Birger, C., Thorvaldsdóttir, H., Ghandi, M., Mesirov, J.P., Tamayo, P., The molecular signatures database hallmark gene set collection. Cell Syst. 1 (2015), 417–425, 10.1016/j.cels.2015.12.004.
Lin, T., Lin, Y., P53 switches off pluripotency on differentiation. Stem Cell Res. Ther., 8, 2017, 44, 10.1186/s13287-017-0498-1.
Liu, J., Lichtenberg, T., Hoadley, K.A., Poisson, L.M., Lazar, A.J., Cherniack, A.D., Kovatich, A.J., Benz, C.C., Levine, D.A., Lee, A.V., et al. An integrated TCGA pan-cancer clinical data resource to drive high-quality survival outcome analytics. Cell 173 (2018), 400–416.e11, 10.1016/j.cell.2018.02.052.
Löffler, M.W., Mohr, C., Bichmann, L., Freudenmann, L.K., Walzer, M., Schroeder, C.M., Trautwein, N., Hilke, F.J., Zinser, R.S., Mühlenbruch, L., et al. Multi-omics discovery of exome-derived neoantigens in hepatocellular carcinoma. Genome Med. 11 (2019), 28–36, 10.1186/s13073-019-0636-8.
Loizou, E., Banito, A., Livshits, G., Ho, Y.J., Koche, R.P., Sánchez-Rivera, F.J., Mayle, A., Chen, C.C., Kinalis, S., Bagger, F.O., et al. A gain-of-function p53-mutant oncogene promotes cell fate plasticity and myeloid leukemia through the pluripotency factor foxh1. Cancer Discov. 9 (2019), 962–979, 10.1158/2159-8290.CD-18-1391.
GTEx Consortium Thomas, J., Salvatore, M., Phillips, R., Lo, E., Shad, S., Hasz, R., Walters, G., Garcia, F., Young, N., et al. The genotype-tissue expression (GTEx) project. Nat. Genet. 45 (2013), 580–585, 10.1038/ng.2653.
Lu, X., Sachs, F., Ramsay, L., Jacques, P.É., Göke, J., Bourque, G., Ng, H.H., The retrovirus HERVH is a long noncoding RNA required for human embryonic stem cell identity. Nat. Struct. Mol. Biol. 21 (2014), 423–425, 10.1038/nsmb.2799.
Ma, J., Zhao, J., Lu, J., Wang, P., Feng, H., Zong, Y., Ou, B., Zheng, M., Lu, A., Cadherin-12 enhances proliferation in colorectal cancer cells and increases progression by promoting EMT. Tumour Biol. 37 (2016), 9077–9088, 10.1007/s13277-015-4555-z.
Macrae, T., Sargeant, T., Lemieux, S., Hébert, J., Deneault, E., Sauvageau, G., RNA-Seq reveals spliceosome and proteasome genes as most consistent transcripts in human cancer cells. PLoS One, 8, 2013, e72884, 10.1371/journal.pone.0072884.
Madsen, R.R., PI3K in stemness regulation: from development to cancer. Biochem. Soc. Trans. 48 (2020), 301–315, 10.1042/BST20190778.
Maiga, A., Lemieux, S., Pabst, C., Lavallée, V.P., Bouvier, M., Sauvageau, G., Hébert, J., Transcriptome analysis of G protein-coupled receptors in distinct genetic subgroups of acute myeloid leukemia: identification of potential disease-specific targets. Blood Cancer J., 6, 2016, e431, 10.1038/bcj.2016.36.
Malta, T.M., Sokolov, A., Gentles, A.J., Burzykowski, T., Poisson, L., Weinstein, J.N., Kamińska, B., Huelsken, J., Omberg, L., Gevaert, O., et al. Machine learning identifies stemness features associated with oncogenic dedifferentiation. Cell 173 (2018), 338–354.e15, 10.1016/j.cell.2018.03.034.
Marçais, G., Kingsford, C., A fast, lock-free approach for efficient parallel counting of occurrences of k-mers. Bioinformatics 27 (2011), 764–770, 10.1093/bioinformatics/btr011.
McLaughlin, R.N., Young, J.M., Yang, L., Neme, R., Wichman, H.A., Malik, H.S., Positive selection and multiple losses of the LINE-1-derived L1TD1 gene in mammals suggest a dual role in genome defense and pluripotency. PLoS Genet., 10, 2014, e1004531, 10.1371/journal.pgen.1004531.
Merkle, F.T., Ghosh, S., Kamitaki, N., Mitchell, J., Avior, Y., Mello, C., Kashin, S., Mekhoubad, S., Ilic, D., Charlton, M., et al. Human pluripotent stem cells recurrently acquire and expand dominant negative P53 mutations. Nature 545 (2017), 229–233, 10.1038/nature22312.
Mi, H., Ebert, D., Muruganujan, A., Mills, C., Albou, L.-P., Mushayamaha, T., Thomas, P.D., PANTHER version 16: a revised family classification, tree-based classification tool, enhancer regions and extensive API. Nucleic Acids Res. 49 (2021), D394–D403, 10.1093/nar/gkaa1106.
Miranda, A., Hamilton, P.T., Zhang, A.W., Pattnaik, S., Becht, E., Mezheyeuski, A., Bruun, J., Micke, P., de Reynies, A., Nelson, B.H., Cancer stemness, intratumoral heterogeneity, and immune response across cancers. Proc. Natl. Acad. Sci. USA 116 (2019), 9020–9029, 10.1073/pnas.1818210116.
Närvä, E., Rahkonen, N., Emani, M.R., Lund, R., Pursiheimo, J.P., Nästi, J., Autio, R., Rasool, O., Denessiouk, K., Lähdesmäki, H., et al. RNA-binding protein L1TD1 interacts with LIN28 via RNA and is required for human embryonic stem cell self-renewal and cancer cell proliferation. Stem Cell. 30 (2012), 452–460, 10.1002/stem.1013.
Di Nicola, M., Carlo-Stella, C., Magni, M., Milanesi, M., Longoni, P.D., Matteucci, P., Grisanti, S., Gianni, A.M., Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood 99 (2002), 3838–3843, 10.1182/blood.V99.10.3838.
Noronha, N., Ehx, G., Meunier, M.-C., Laverdure, J.-P., Thériault, C., Perreault, C., Major multilevel molecular divergence between THP-1 cells from different biorepositories. Int. J. Cancer 147 (2020), 2000–2006, 10.1002/ijc.32967.
Ogishi, M., Yotsuyanagi, H., Quantitative prediction of the landscape of T cell epitope immunogenicity in sequence space. Front. Immunol., 10, 2019, 827, 10.3389/fimmu.2019.00827.
Durette, C., Daouda, T., Granados, D.P., Pearson, H., Bonneil, E., Courcelles, M., Rodenbrock, A., Laverdure, J.P., Côté, C., Mader, S., et al. MHC class I – associated peptides derive from selective regions of the human genome. J. Clin. Invest. 126 (2016), 4690–4701, 10.1172/JCI88590.defective.
Pellagatti, A., Armstrong, R.N., Steeples, V., Sharma, E., Repapi, E., Singh, S., Sanchi, A., Radujkovic, A., Horn, P., Dolatshad, H., et al. Impact of spliceosome mutations on RNA splicing in myelodysplasia: dysregulated genes/pathways and clinical associations. Blood 132 (2018), 1225–1240, 10.1182/blood-2018-04-843771.
Pelullo, M., Zema, S., Nardozza, F., Checquolo, S., Screpanti, I., Bellavia, D., Wnt, Notch, and TGF-β pathways impinge on hedgehog signaling complexity: an open window on cancer. Front. Genet. 10 (2019), 711–716, 10.3389/fgene.2019.00711.
Petroni, G., Buqué, A., Zitvogel, L., Kroemer, G., Galluzzi, L., Immunomodulation by targeted anticancer agents. Cancer Cell 39 (2021), 310–345, 10.1016/j.ccell.2020.11.009.
Poli, V., Fagnocchi, L., Fasciani, A., Cherubini, A., Mazzoleni, S., Ferrillo, S., Miluzio, A., Gaudioso, G., Vaira, V., Turdo, A., et al. MYC-driven epigenetic reprogramming favors the onset of tumorigenesis by inducing a stem cell-like state. Nat. Commun., 9, 2018, 1024, 10.1038/s41467-018-03264-2.
Prensner, J.R., Iyer, M.K., Balbin, O.A., Dhanasekaran, S.M., Cao, Q., Brenner, J.C., Laxman, B., Asangani, I.A., Grasso, C.S., Kominsky, H.D., et al. Transcriptome sequencing across a prostate cancer cohort identifies PCAT-1, an unannotated lincRNA implicated in disease progression. Nat. Biotechnol. 29 (2011), 742–749, 10.1038/nbt.1914.
Reinhard, K., Rengstl, B., Oehm, P., Michel, K., Billmeier, A., Hayduk, N., Klein, O., Kuna, K., Ouchan, Y., Wöll, S., et al. An RNA vaccine drives expansion and efficacy of claudin-CAR-T cells against solid tumors. Science 367 (2020), 446–453, 10.1126/science.aay5967.
Van Rhenen, A., Feller, N., Kelder, A., Westra, A.H., Rombouts, E., Zweegman, S., Van Der Pol, M.A., Waisfisz, Q., Ossenkoppele, G.J., Schuurhuis, G.J., High stem cell frequency in acute myeloid leukemia at diagnosis predicts high minimal residual disease and poor survival. Clin. Cancer Res. 11 (2005), 6520–6527, 10.1158/1078-0432.CCR-05-0468.
Ruiz Cuevas, M.V., Hardy, M.P., Hollý, J., Bonneil, É., Durette, C., Courcelles, M., Lanoix, J., Côté, C., Staudt, L.M., Lemieux, S., et al. Most non-canonical proteins uniquely populate the proteome or immunopeptidome. Cell Rep., 34, 2021, 108815, 10.1016/j.celrep.2021.108815.
Sahin, U., Oehm, P., Derhovanessian, E., Jabulowsky, R.A., Vormehr, M., Gold, M., Maurus, D., Schwarck-Kokarakis, D., Kuhn, A.N., Omokoko, T., et al. An RNA vaccine drives immunity in checkpoint-inhibitor-treated melanoma. Nature 585 (2020), 107–112, 10.1038/s41586-020-2537-9.
Salmon, H., Idoyaga, J., Rahman, A., Leboeuf, M., Remark, R., Jordan, S., Casanova-Acebes, M., Khudoynazarova, M., Agudo, J., Tung, N., et al. Expansion and activation of CD103+ dendritic cell progenitors at the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. Immunity 44 (2016), 924–938, 10.1016/j.immuni.2016.03.012.
Sanchez-Vega, F., Mina, M., Armenia, J., Chatila, W.K., Luna, A., La, K.C., Dimitriadoy, S., Liu, D.L., Kantheti, H.S., Saghafinia, S., et al. Oncogenic signaling pathways in the cancer genome Atlas. Cell 173 (2018), 321–337.e10, 10.1016/j.cell.2018.03.035.
Schumacher, T.N., Scheper, W., Kvistborg, P., Cancer neoantigens. Annu. Rev. Immunol. 37 (2019), 173–200, 10.1146/annurev-immunol-042617-053402.
Schuster, H., Peper, J.K., Bösmüller, H.C., Röhle, K., Backert, L., Bilich, T., Ney, B., Löffler, M.W., Kowalewski, D.J., Trautwein, N., et al. The immunopeptidomic landscape of ovarian carcinomas. Proc. Natl. Acad. Sci. USA 114 (2017), E9942–E9951, 10.1073/pnas.1707658114.
Shan, X., Roberts, C., Kim, E.J., Brenner, A., Grant, G., Percec, I., Transcriptional and cell cycle alterations mark aging of primary human adipose-derived stem cells. Stem Cell. 35 (2017), 1392–1401, 10.1002/stem.2592.
Shen, W.-C., Lai, Y.-C., Li, L.-H., Liao, K., Lai, H.-C., Kao, S.-Y., Wang, J., Chuong, C.-M., Hung, S.-C., Methylation and PTEN activation in dental pulp mesenchymal stem cells promotes osteogenesis and reduces oncogenesis. Nat. Commun., 10, 2019, 2226, 10.1038/s41467-019-10197-x.
Smith, B.A., Balanis, N.G., Nanjundiah, A., Sheu, K.M., Tsai, B.L., Zhang, Q., Park, J.W., Thompson, M., Huang, J., Witte, O.N., Graeber, T.G., A human adult stem cell signature marks aggressive variants across epithelial cancers. Cell Rep. 24 (2018), 3353–3366.e5, 10.1016/j.celrep.2018.08.062.
Spranger, S., Gajewski, T.F., Impact of oncogenic pathways on evasion of antitumour immune responses. Nat. Rev. Cancer 18 (2018), 139–147, 10.1038/nrc.2017.117.
Spranger, S., Dai, D., Horton, B., Gajewski, T.F., Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell 31 (2017), 711–723.e4, 10.1016/j.ccell.2017.04.003.
Stewart, M.H., Bossé, M., Chadwick, K., Menendez, P., Bendall, S.C., Bhatia, M., Clonal isolation of hESCs reveals heterogeneity within the pluripotent stem cell compartment. Nat. Methods 3 (2006), 807–815, 10.1038/nmeth939.
Stine, Z.E., Walton, Z.E., Altman, B.J., Hsieh, A.L., Dang, C.V., MYC, metabolism, and cancer. Cancer Discov. 5 (2015), 1024–1039, 10.1158/2159-8290.CD-15-0507.
Szolek, A., Schubert, B., Mohr, C., Sturm, M., Feldhahn, M., Kohlbacher, O., OptiType: precision HLA typing from next-generation sequencing data. Bioinformatics 30 (2014), 3310–3316, 10.1093/bioinformatics/btu548.
Thorsson, V., Gibbs, D.L., Brown, S.D., Wolf, D., Bortone, D.S., Ou Yang, T.H., Porta-Pardo, E., Gao, G.F., Plaisier, C.L., Eddy, J.A., et al. The immune landscape of cancer. Immunity 48 (2018), 812–830.e14, 10.1016/j.immuni.2018.03.023.
Vasileiou, S., Lulla, P.D., Tzannou, I., Watanabe, A., Kuvalekar, M., Callejas, W.L., Bilgi, M., Wang, T., Wu, M.J., Kamble, R., et al. T-cell therapy for lymphoma using nonengineered multiantigen-targeted T cells is safe and produces durable clinical effects. J. Clin. Oncol. 39 (2021), 1415–1425, 10.1200/jco.20.02224.
Vogel, C., Marcotte, E.M., Absolute abundance for the masses. Nat. Biotechnol. 27 (2009), 825–826, 10.1038/nbt0909-825.
Wang, J.F., She, L., Su, B.H., Ding, L.C., Zheng, F.F., Zheng, D.L., Lu, Y.G., CDH12 promotes the invasion of salivary adenoid cystic carcinoma. Oncol. Rep. 26 (2011), 101–108, 10.3892/or.2011.1286.
Wang, L., Su, Y., Huang, C., Yin, Y., Zhu, J., Knupp, A., Chu, A., Tang, Y., FOXH1 is regulated by NANOG and LIN28 for early-stage reprogramming. Sci. Rep. 9 (2019), 16443–16448, 10.1038/s41598-019-52861-8.
Wang, Z.-X., Kueh, J.L.L., Teh, C.H.-L., Rossbach, M., Lim, L., Li, P., Wong, K.-Y., Lufkin, T., Robson, P., Stanton, L.W., Zfp206 is a transcription factor that controls pluripotency of embryonic stem cells. Stem Cell. 25 (2007), 2173–2182, 10.1634/stemcells.2007-0085.
Bartolo, D., Guo, J.U., Coding functions of “noncoding” RNAs. Science 367 (2020), 1075–1076, 10.1126/science.aba5319.
Wu, T.D., Nacu, S., Fast and SNP-tolerant detection of complex variants and splicing in short reads. Bioinformatics 26 (2010), 873–881, 10.1093/bioinformatics/btq057.
Yaddanapudi, K., Mitchell, R.A., Putty, K., Willer, S., Sharma, R.K., Yan, J., Bodduluri, H., Eaton, J.W., Vaccination with embryonic stem cells protects against lung cancer: is a broad-spectrum prophylactic vaccine against cancer possible?. PLoS One, 7, 2012, e42289, 10.1371/journal.pone.0042289.
Yewdell, J.W., Designing CD8+ T cell vaccines: it's not rocket science (yet). Curr. Opin. Immunol. 22 (2010), 402–410, 10.1016/j.coi.2010.04.002.
Yu, H.B., Kunarso, G., Hong, F.H., Stanton, L.W., Zfp206, Oct4, and Sox2 are integrated components of a transcriptional regulatory network in embryonic stem cells. J. Biol. Chem. 284 (2009), 31327–31335, 10.1074/jbc.M109.016162.
Zhang, H.L., Wang, P., Lu, M.Z., Zhang, S.D., Zheng, L., c-Myc maintains the self-renewal and chemoresistance properties of colon cancer stem cells. Oncol. Lett. 17 (2019), 4487–4493, 10.3892/ol.2019.10081.
Zhang, J., Nuebel, E., Daley, G.Q., Koehler, C.M., Teitell, M.A., Metabolic regulation in pluripotent stem cells during reprogramming and self-renewal. Cell Stem Cell 11 (2012), 589–595, 10.1016/j.stem.2012.10.005.
Zhang, J., Ratanasirintrawoot, S., Chandrasekaran, S., Wu, Z., Ficarro, S.B., Yu, C., Ross, C.A., Cacchiarelli, D., Xia, Q., Seligson, M., et al. LIN28 regulates stem cell metabolism and conversion to primed pluripotency. Cell Stem Cell 19 (2016), 66–80, 10.1016/j.stem.2016.05.009.
Zhao, Q., Laverdure, J.-P., Lanoix, J., Durette, C., Côté, C., Bonneil, É., Laumont, C.M., Gendron, P., Vincent, K., Courcelles, M., et al. Proteogenomics uncovers a vast repertoire of shared tumor-specific antigens in ovarian cancer. Cancer Immunol. Res. 8 (2020), 544–555, 10.1158/2326-6066.cir-19-0541.
Zhao, S., Zhu, W., Xue, S., Han, D., Testicular defense systems: immune privilege and innate immunity. Cell. Mol. Immunol. 11 (2014), 428–437, 10.1038/cmi.2014.38.
Zheng, K., Wu, X., Kaestner, K.H., Wang, P.J., The pluripotency factor LIN28 marks undifferentiated spermatogonia in mouse. BMC Dev. Biol. 9 (2009), 38–41, 10.1186/1471-213X-9-38.
Zhou, X., Guo, X., Chen, M., Xie, C., Jiang, J., HIF-3α promotes metastatic phenotypes in pancreatic cancer by transcriptional regulation of the RhoC-ROCK1 signaling pathway. Mol. Cancer Res. 16 (2018), 124–134, 10.1158/1541-7786.MCR-17-0256.