High Somatic Mutation and Neoantigen Burden Do Not Correlate with Decreased Progression-Free Survival in HCC Patients not Undergoing Immunotherapy
Abstract
:1. Introduction
2. Results
2.1. Tumor Mutational Burden and Survival
2.2. Neoantigen Prediction
2.3. Correlation between Neoantigens and Survival
2.4. Impact of HLA on the Survival
2.5. Impact of Tumor Microenvironment of Survival
2.6. Correlation of Neoantigens and Tumor Microenvironment
2.7. Sequence Homology Analysis with Known Epitopes
3. Discussion
4. Materials and Methods
4.1. Clinical Cohorts and Outcome Assessments
4.2. Neopeptide Prediction and DAI Analysis
4.3. Epitope Prediction and Sequence Homology Analysis
4.4. Assessment of Tumor Infiltrating Immune Cells
4.5. Statistical Analysis
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Conflicts of Interest
References
- Greenman, C.; Stephens, P.; Smith, R.; Dalgliesh, G.L.; Hunter, C.; Bignell, G.; Davies, H.; Teague, J.; Butler, A.; Stevens, C.; et al. Patterns of somatic mutation in human cancer genomes. Nature 2007, 446, 153–158. [Google Scholar] [CrossRef]
- Stratton, M.R. Exploring the genomes of cancer cells: progress and promise. Science 2011, 331, 1553–1558. [Google Scholar] [CrossRef] [PubMed]
- Lennerz, V.; Fatho, M.; Gentilini, C.; Frye, R.A.; Lifke, A.; Ferel, D.; Wolfel, C.; Huber, C.; Wolfel, T. The response of autologous T cells to a human melanoma is dominated by mutated neoantigens. Proc. Natl. Acad. Sci. USA 2005, 102, 16013–16018. [Google Scholar]
- Robbins, P.F.; Lu, Y.C.; El-Gamil, M.; Li, Y.F.; Gross, C.; Gartner, J.; Lin, J.C.; Teer, J.K.; Cliften, P.; Tycksen, E.; et al. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat. Med. 2013, 19, 747–752. [Google Scholar] [CrossRef] [PubMed]
- Wick, D.A.; Webb, J.R.; Nielsen, J.S.; Martin, S.D.; Kroeger, D.R.; Milne, K.; Castellarin, M.; Twumasi-Boateng, K.; Watson, P.H.; Holt, R.A.; et al. Surveillance of the tumor mutanome by T cells during progression from primary to recurrent ovarian cancer. Clin. Cancer Res. 2014, 20, 1125–1134. [Google Scholar] [CrossRef] [PubMed]
- Schumacher, T.N.; Schreiber, R.D. Neoantigens in cancer immunotherapy. Science 2015, 348, 69–74. [Google Scholar] [CrossRef]
- Yarchoan, M.; Johnson, B.A., III; Lutz, E.R.; Laheru, D.A.; Jaffee, E.M. Targeting neoantigens to augment antitumour immunity. Nat. Rev. Cancer 2017, 17, 569. [Google Scholar] [CrossRef]
- Brown, S.D.; Warren, R.L.; Gibb, E.A.; Martin, S.D.; Spinelli, J.J.; Nelson, B.H.; Holt, R.A. Neo-antigens predicted by tumor genome meta-analysis correlate with increased patient survival. Genome Res. 2014, 24, 743–750. [Google Scholar] [CrossRef]
- Lu, Y.C.; Yao, X.; Crystal, J.S.; Li, Y.F.; El-Gamil, M.; Gross, C.; Davis, L.; Dudley, M.E.; Yang, J.C.; Samuels, Y.; et al. Efficient identification of mutated cancer antigens recognized by T cells associated with durable tumor regressions. Clin Cancer Res. 2014, 20, 3401–3410. [Google Scholar] [CrossRef]
- Ott, P.A.; Bang, Y.J.; Piha-Paul, S.A.; Razak, A.R.A.; Bennouna, J.; Soria, J.C.; Rugo, H.S.; Cohen, R.B.; O’Neil, B.H.; Mehnert, J.M.; et al. T-cell-inflamed gene-expression profile, programmed death ligand 1 expression, and tumor mutational burden predict efficacy in patients treated with pembrolizumab across 20 cancers: Keynote-028. J. Clin. Oncol. 2019, 37, 318–327. [Google Scholar] [CrossRef]
- Yarchoan, M.; Hopkins, A.; Jaffee, E.M. Tumor mutational burden and response rate to PD-1 inhibition. N. Engl. J. Med. 2017, 377, 2500–2501. [Google Scholar] [CrossRef]
- Miao, D.; Margolis, C.A.; Vokes, N.I.; Liu, D.; Taylor-Weiner, A.; Wankowicz, S.M.; Adeegbe, D.; Keliher, D.; Schilling, B.; Tracy, A.; et al. Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors. Nat. Genet. 2018, 50, 1271–1281. [Google Scholar] [CrossRef]
- Matsushita, H.; Sato, Y.; Karasaki, T.; Nakagawa, T.; Kume, H.; Ogawa, S.; Homma, Y.; Kakimi, K. Neoantigen load, antigen presentation machinery, and immune signatures determine prognosis in clear cell renal cell carcinoma. Cancer Immunol. Res. 2016, 4, 463–471. [Google Scholar] [CrossRef]
- Luen, S.; Virassamy, B.; Savas, P.; Salgado, R.; Loi, S. The genomic landscape of breast cancer and its interaction with host immunity. Breast 2016, 29, 241–250. [Google Scholar] [CrossRef]
- Choudhury, N.J.; Kiyotani, K.; Yap, K.L.; Campanile, A.; Antic, T.; Yew, P.Y.; Steinberg, G.; Park, J.H.; Nakamura, Y.; O’Donnell, P.H. Low T-cell receptor diversity, high somatic mutation burden, and high neoantigen load as predictors of clinical outcome in muscle-invasive bladder cancer. Eur. Urol. Focus. 2016, 2, 445–452. [Google Scholar] [CrossRef]
- Feng, L.; Qian, H.; Yu, X.; Liu, K.; Xiao, T.; Zhang, C.; Kuang, M.; Cheng, S.; Li, X.; Wan, J.; et al. Heterogeneity of tumor-infiltrating lymphocytes ascribed to local immune status rather than neoantigens by multi-omics analysis of glioblastoma multiforme. Sci. Rep. 2017, 7, 6968. [Google Scholar] [CrossRef]
- Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S.; et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015, 348, 124–128. [Google Scholar] [CrossRef]
- Snyder, A.; Makarov, V.; Merghoub, T.; Yuan, J.; Zaretsky, J.M.; Desrichard, A.; Walsh, L.A.; Postow, M.A.; Wong, P.; Ho, T.S.; et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J Med. 2014, 371, 2189–2199. [Google Scholar] [CrossRef]
- Ock, C.Y.; Hwang, J.E.; Keam, B.; Kim, S.B.; Shim, J.J.; Jang, H.J.; Park, S.; Sohn, B.H.; Cha, M.; Ajani, J.A.; et al. Genomic landscape associated with potential response to anti-CTLA-4 treatment in cancers. Genomic landscape associated with potential response to anti-CTLA-4 treatment in cancers. Nat. Commun. 2017, 8, 1050. [Google Scholar] [CrossRef]
- Karpanen, T.; Olweus, J. The potential of donor T-Cell repertoires in neoantigen-targeted cancer immunotherapy. Front Immunol. 2017, 8, 1718. [Google Scholar] [CrossRef]
- Parkhurst, M.; Gros, A.; Pasetto, A.; Prickett, T.; Crystal, J.S.; Robbins, P.; Rosenberg, S.A. Isolation of T-cell receptors specifically reactive with mutated tumor-associated antigens from tumor-infiltrating lymphocytes based on CD137 expression. Clin. Cancer Res. 2017, 23, 2491–2505. [Google Scholar] [CrossRef] [PubMed]
- Seliktar-Ofir, S.; Merhavi-Shoham, E.; Itzhaki, O.; Yunger, S.; Markel, G.; Schachter, J.; Besser, M.J. Selection of shared and neoantigen-reactive T cells for adoptive cell therapy based on CD137 separation. Front Immunol. 2017, 8, 1211. [Google Scholar] [CrossRef] [PubMed]
- Sahin, U.; Derhovanessian, E.; Miller, M.; Kloke, B.P.; Simon, P.; Lower, M.; Bukur, V.; Tadmor, A.D.; Luxemburger, U.; Schrors, B.; et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 2017, 547, 222–226. [Google Scholar] [CrossRef] [PubMed]
- Ott, P.A.; Hu, Z.; Keskin, D.B.; Shukla, S.A.; Sun, J.; Bozym, D.J.; Zhang, W.; Luoma, A.; Giobbie-Hurder, A.; Peter, L.; et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 2017, 547, 217–221. [Google Scholar] [CrossRef] [PubMed]
- Guo, Y.; Lei, K.; Tang, L. Neoantigen vaccine delivery for personalized anticancer immunotherapy. Front Immunol. 2018, 9, 1499. [Google Scholar] [CrossRef] [PubMed]
- Aurisicchio, L.; Pallocca, M.; Ciliberto, G.; Palombo, F. The perfect personalized cancer therapy: cancer vaccines against neoantigens. J. Exp. Clin. Cancer Res. 2018, 37, 86. [Google Scholar] [CrossRef]
- Luksza, M.; Riaz, N.; Makarov, V.; Balachandran, V.P.; Hellmann, M.D.; Solovyov, A.; Rizvi, N.A.; Merghoub, T.; Levine, A.J.; Chan, T.A.; et al. A neoantigen fitness model predicts tumour response to checkpoint blockade immunotherapy. Nature 2017, 551, 517–520. [Google Scholar] [CrossRef]
- Ghorani, E.; Rosenthal, R.; McGranahan, N.; Reading, J.L.; Lynch, M.; Peggs, K.S.; Swanton, C.; Quezada, S.A. Differential binding affinity of mutated peptides for MHC class I is a predictor of survival in advanced lung cancer and melanoma. Ann. Oncol. 2018, 29, 271–279. [Google Scholar] [CrossRef] [PubMed]
- Rech, A.J.; Balli, D.; Mantero, A.; Ishwaran, H.; Nathanson, K.L.; Stanger, B.Z.; Vonderheide, R.H. Tumor immunity and survival as a function of alternative neopeptides in human cancer. Cancer Immunol. Res. 2018, 6, 276–287. [Google Scholar] [CrossRef]
- Balachandran, V.P.; Luksza, M.; Zhao, J.N.; Makarov, V.; Moral, J.A.; Remark, R.; Herbst, B.; Askan, G.; Bhanot, U.; Senbabaoglu, Y.; et al. Identification of unique neoantigen qualities in long-term survivors of pancreatic cancer. Nature 2017, 551, 512–516. [Google Scholar] [CrossRef]
- Fujimoto, A.; Furuta, M.; Totoki, Y.; Tsunoda, T.; Kato, M.; Shiraishi, Y.; Tanaka, H.; Taniguchi, H.; Kawakami, Y.; Ueno, M.; et al. Whole-genome mutational landscape and characterization of noncoding and structural mutations in liver cancer. Nat. Genet. 2016, 48, 500–509. [Google Scholar] [CrossRef]
- Loffler, M.W.; Mohr, C.; Bichmann, L.; Freudenmann, L.K.; Walzer, M.; Schroeder, C.M.; Trautwein, N.; Hilke, F.J.; Zinser, R.S.; Muhlenbruch, L.; et al. Multi-omics discovery of exome-derived neoantigens in hepatocellular carcinoma. Genome Med. 2019, 11, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Petrizzo, A.; Tagliamonte, M.; Mauriello, A.; Costa, V.; Aprile, M.; Esposito, R.; Caporale, A.; Luciano, A.; Arra, C.; Tornesello, M.L.; et al. Unique true predicted neoantigens (TPNAs) correlates with anti-tumor immune control in HCC patients. J. Transl. Med. 2018, 16, 286. [Google Scholar] [CrossRef] [PubMed]
- Fritsch, E.F.; Rajasagi, M.; Ott, P.A.; Brusic, V.; Hacohen, N.; Wu, C.J. HLA-binding properties of tumor neoepitopes in humans. Cancer Immunol. Res. 2014, 2, 522–529. [Google Scholar] [CrossRef] [PubMed]
- Schumacher, T.N.; Scheper, W.; Kvistborg, P. Cancer Neoantigens. Annu. Rev. Immunol. 2019, 37, 173–200. [Google Scholar] [CrossRef] [PubMed]
- Samstein, R.M.; Lee, C.H.; Shoushtari, A.N.; Hellmann, M.D.; Shen, R.; Janjigian, Y.Y.; Barron, D.A.; Zehir, A.; Jordan, E.J.; Omuro, A.; et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat. Genet. 2019, 51, 202–206. [Google Scholar] [CrossRef]
- Miller, A.; Asmann, Y.; Cattaneo, L.; Braggio, E.; Keats, J.; Auclair, D.; Lonial, S.; Russell, S.J.; Stewart, A.K. High somatic mutation and neoantigen burden are correlated with decreased progression-free survival in multiple myeloma. Blood Cancer J. 2017, 7, e612. [Google Scholar]
- European Association for the Study of the Liver; European Organisation for Research and Treatment of Cancer. EASL-EORTC clinical practice guidelines: Management of hepatocellular carcinoma. J. Hepatol. 2012, 56, 908–943. [Google Scholar] [CrossRef] [Green Version]
- Duan, F.; Duitama, J.; Al, S.S.; Ayres, C.M.; Corcelli, S.A.; Pawashe, A.P.; Blanchard, T.; McMahon, D.; Sidney, J.; Sette, A.; et al. Genomic and bioinformatic profiling of mutational neoepitopes reveals new rules to predict anticancer immunogenicity. J. Exp. Med. 2014, 211, 2231–2248. [Google Scholar] [CrossRef]
Group | Characteristics |
---|---|
sex | |
male | 61.50% |
female | 38.50% |
age | 18–90 years (average 61.5 years) |
etiology | |
HBV | 19.20% |
HCV | 36.10% |
Non viral | 44.60% |
stage | |
I | 33% |
II | 19.20% |
III | 34.60% |
IV | 2.30% |
N/A | 10.80% |
Treatment | |
RF | 4.60% |
CE | 11.50% |
RE | 3.80% |
N/A | 80% |
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Mauriello, A.; Zeuli, R.; Cavalluzzo, B.; Petrizzo, A.; Tornesello, M.L.; Buonaguro, F.M.; Ceccarelli, M.; Tagliamonte, M.; Buonaguro, L. High Somatic Mutation and Neoantigen Burden Do Not Correlate with Decreased Progression-Free Survival in HCC Patients not Undergoing Immunotherapy. Cancers 2019, 11, 1824. https://doi.org/10.3390/cancers11121824
Mauriello A, Zeuli R, Cavalluzzo B, Petrizzo A, Tornesello ML, Buonaguro FM, Ceccarelli M, Tagliamonte M, Buonaguro L. High Somatic Mutation and Neoantigen Burden Do Not Correlate with Decreased Progression-Free Survival in HCC Patients not Undergoing Immunotherapy. Cancers. 2019; 11(12):1824. https://doi.org/10.3390/cancers11121824
Chicago/Turabian StyleMauriello, Angela, Roberta Zeuli, Beatrice Cavalluzzo, Annacarmen Petrizzo, Maria Lina Tornesello, Franco M. Buonaguro, Michele Ceccarelli, Maria Tagliamonte, and Luigi Buonaguro. 2019. "High Somatic Mutation and Neoantigen Burden Do Not Correlate with Decreased Progression-Free Survival in HCC Patients not Undergoing Immunotherapy" Cancers 11, no. 12: 1824. https://doi.org/10.3390/cancers11121824
APA StyleMauriello, A., Zeuli, R., Cavalluzzo, B., Petrizzo, A., Tornesello, M. L., Buonaguro, F. M., Ceccarelli, M., Tagliamonte, M., & Buonaguro, L. (2019). High Somatic Mutation and Neoantigen Burden Do Not Correlate with Decreased Progression-Free Survival in HCC Patients not Undergoing Immunotherapy. Cancers, 11(12), 1824. https://doi.org/10.3390/cancers11121824