Mutant MHC class II epitopes drive therapeutic immune responses to cancer (original) (raw)
References
- Castle, J. C. et al. Exploiting the mutanome for tumor vaccination. Cancer Res. 72, 1081–1091 (2012).
Article CAS Google Scholar - Castle, J. C. et al. Immunomic, genomic and transcriptomic characterization of CT26 colorectal carcinoma. BMC Genomics 15, 190 (2014).
Article Google Scholar - Holtkamp, S. et al. Modification of antigen-encoding RNA increases stability, translational efficacy, and T-cell stimulatory capacity of dendritic cells. Blood 108, 4009–4017 (2006).
Article CAS Google Scholar - Kreiter, S. et al. Increased antigen presentation efficiency by coupling antigens to MHC class I trafficking signals. J. Immunol. 180, 309–318 (2008).
Article CAS Google Scholar - Kuhn, A. N. et al. Phosphorothioate cap analogs increase stability and translational efficiency of RNA vaccines in immature dendritic cells and induce superior immune responses in vivo. Gene Ther. 17, 961–971 (2010).
Article CAS Google Scholar - Bloom, M. B. et al. Identification of tyrosinase-related protein 2 as a tumor rejection antigen for the B16 melanoma. J. Exp. Med. 185, 453–459 (1997).
Article CAS Google Scholar - Schumacher, T. et al. A vaccine targeting mutant IDH1 induces antitumour immunity. Nature 512, 324–327 (2014).
Article ADS CAS Google Scholar - Tran, E. et al. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science 344, 641–645 (2014).
Article ADS CAS Google Scholar - Britten, C. M. et al. The regulatory landscape for actively personalized cancer immunotherapies. Nature Biotechnol. 31, 880–882 (2013).
Article CAS Google Scholar - Gerlinger, M. et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 366, 883–892 (2012).
Article CAS Google Scholar - Koebel, C. M. et al. Adaptive immunity maintains occult cancer in an equilibrium state. Nature 450, 903–907 (2007).
Article ADS CAS Google Scholar - Matsushita, H. et al. Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting. Nature 482, 400–404 (2012).
Article ADS CAS Google Scholar - Robbins, P. F. et al. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nature Med. 19, 747–752 (2013).
Article CAS Google Scholar - van Rooij, N. et al. Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. J. Clin. Oncol. 31, e439–e442 (2013).
Article Google Scholar - Shen, Z. et al. Cloned dendritic cells can present exogenous antigens on both MHC class I and class II molecules. J. Immunol. 158, 2723–2720 (1997).
CAS PubMed Google Scholar - Schoenberger, S. P. et al. T-cell help for cytotoxic T lymphocytes is mediated by CD40–CD40L interactions. Nature 393, 480–483 (1998).
Article ADS CAS Google Scholar - Linnemann, C. et al. High-throughput epitope discovery reveals frequent recognition of neo-antigens by CD4+ T cells in human melanoma. Nature Med. 21, 81–85 (2015).
Article CAS Google Scholar - Arnold, P. Y. et al. The majority of immunogenic epitopes generate CD4+ T cells that are dependent on MHC class II-bound peptide-flanking residues. J. Immunol. 169, 739–749 (2002).
Article CAS Google Scholar - Wolfel, T. et al. A p16INK4a-insensitive CDK4 mutant targeted by cytolytic T lymphocytes in a human melanoma. Science 269, 1281–1284 (1995).
Article ADS CAS Google Scholar - Wang, R. F. et al. Cloning genes encoding MHC class II-restricted antigens: mutated CDC27 as a tumor antigen. Science 284, 1351–1354 (1999).
Article ADS CAS Google Scholar - Lu, Y. C. et al. Mutated PPP1R3B is recognized by T cells used to treat a melanoma patient who experienced a durable complete tumor regression. J. Immunol. 190, 6034–6042 (2013).
Article CAS Google Scholar - Gubin, M. M. et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 515, 577–581 (2014).
Article ADS CAS Google Scholar - Snyder, A. et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 371, 2189–2199 (2014).
Article Google Scholar - Herbst, R. S. et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 515, 563–567 (2014).
Article ADS CAS Google Scholar - Tumeh, P. C. et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515, 568–571 (2014).
Article ADS CAS Google Scholar - Rizvi, N. A. et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348, 124–128 (2015).
Article ADS CAS Google Scholar - Castle, J. C. et al. Mutated tumor alleles are expressed according to their DNA frequency. Sci. Rep. 4, 4743 (2014).
Article Google Scholar - Löwer, M. et al. Confidence-based somatic mutation evaluation and prioritization. PLOS Comput. Biol. 8, e1002714 (2012).
Article Google Scholar - Boegel, S. et al. A catalog of HLA type, HLA expression, and neo-epitope candidates in human cancer cell lines. OncoImmunology 3, e954893 (2014).
Article Google Scholar - Kreiter, S. et al. Intranodal vaccination with naked antigen-encoding RNA elicits potent prophylactic and therapeutic antitumoral immunity. Cancer Res. 70, 9031–9040 (2010).
Article CAS Google Scholar
Acknowledgements
We thank M. Holzmann, A. König, U. Schmitt, R. Roth, C. Worm and N. Krause for technical assistance; L. Ralla, J. Groß, A. Spruß, M. Erdeljan, S. Wöll and C. Rohde for immunohistochemical staining and analysis; C. Paret for sequence validation of mutations; M. Brkic for immunofluorescence staining; S. Witzel and Bodo Tillmann, S. Wurzel and Z. Yildiz for cloning of constructs; S. Kind, M. Mechler, F. Wille, B. Otte and S. Petri for RNA production as well as L. Kranz and colleagues involved in RNA formulation development. We are grateful to B. Kloke, S. Heesch, A. Kuhn, J. Buck, C. Britten and H. Haas for conceptual and technical discussions. Moreover, we would like to thank V. Bukur, J. de Graf and C. Albrecht who supported the next-generation sequencing of samples. Furthermore we like to acknowledge A. Kong for critical reading and A. Orlandini for help with graphic design. The results shown here are in part based on data generated by the TCGA Research Network http://cancergenome.nih.gov/. The study was supported by the CI3 excellence cluster program of the Federal Ministry of Education and Research (BMBF).
Author information
Author notes
- Mathias Vormehr and Niels van de Roemer: These authors contributed equally to this work.
- Özlem Türeci and Ugur Sahin: These authors jointly supervised this work.
Authors and Affiliations
- TRON – Translational Oncology at the University Medical Center of Johannes Gutenberg University, Freiligrathstrasse 12, Mainz, 55131, Germany
Sebastian Kreiter, Mustafa Diken, Martin Löwer, Jan Diekmann, Sebastian Boegel, Barbara Schrörs, Fulvia Vascotto, John C. Castle, Arbel D. Tadmor, Özlem Türeci & Ugur Sahin - Research Center for Immunotherapy (FZI), Langenbeckstrasse 1, Building 708, Mainz, 55131, Germany
Mathias Vormehr, Niels van de Roemer, Christoph Huber & Ugur Sahin - Biopharmaceutical New Technologies (BioNTech) Corporation, An der Goldgrube 12, Mainz, 55131, Germany
Jan Diekmann & Ugur Sahin - La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, 92037, California, USA
Stephen P. Schoenberger
Authors
- Sebastian Kreiter
You can also search for this author inPubMed Google Scholar - Mathias Vormehr
You can also search for this author inPubMed Google Scholar - Niels van de Roemer
You can also search for this author inPubMed Google Scholar - Mustafa Diken
You can also search for this author inPubMed Google Scholar - Martin Löwer
You can also search for this author inPubMed Google Scholar - Jan Diekmann
You can also search for this author inPubMed Google Scholar - Sebastian Boegel
You can also search for this author inPubMed Google Scholar - Barbara Schrörs
You can also search for this author inPubMed Google Scholar - Fulvia Vascotto
You can also search for this author inPubMed Google Scholar - John C. Castle
You can also search for this author inPubMed Google Scholar - Arbel D. Tadmor
You can also search for this author inPubMed Google Scholar - Stephen P. Schoenberger
You can also search for this author inPubMed Google Scholar - Christoph Huber
You can also search for this author inPubMed Google Scholar - Özlem Türeci
You can also search for this author inPubMed Google Scholar - Ugur Sahin
You can also search for this author inPubMed Google Scholar
Contributions
U.S. is principal investigator, conceptualized the study and experimental strategy. S.K., M.V., N.vdR., M.D., J.D., F.V. and U.S. planned and analysed experiments. M.V. and N.vdR. performed experiments. S.K., M.V., M.D., S.P.S., C.H., Ö.T. and U.S. interpreted the data and wrote the manuscript. M.L., S.B., A.D.T. and J.C.C. processed next-generation sequencing data and identified mutations. M.V. and B.S. analysed murine MHC II binding predictions. S.B. analysed TCGA data and human MHC II binding predictions.
Corresponding author
Correspondence toUgur Sahin.
Extended data figures and tables
Extended Data Figure 1 Non synonymous cancer‐associated mutations are frequently immunogenic and pre‐dominantly recognized by CD4+ T cells.
T‐cell responses obtained by vaccinating C57BL/6 mice with antigen‐encoding RNA in the B16F10 tumour model (n = 5). Left, prevalence of non‐immunogenic, MHC-class-I- or class-II-restricted mutated epitopes. Right, detection and typing of mutation‐specific T cells (individual epitopes shown in Extended Data Table 1).
Extended Data Figure 2 Mutant epitope-specific T cells induced by RNA vaccination control tumour growth.
a, Splenocytes of mice (n = 5) vaccinated with B16‐M30 RNA were tested by ELISpot for recognition of mutated peptides as compared to the corresponding wild‐type (B16‐WT30) sequence. Right, testing of truncated variants of B16‐M30 (mean + s.e.m.). b, Mean ± s.e.m. tumour growth (left) and survival (right) of C57BL/6 mice (n = 10) inoculated s.c. with B16F10 and left untreated (control) or injected i.v. with irrelevant RNA. c, Lungs of B16F10‐Luc tumour bearing mice shown in Fig. 2b (day 27 after tumour inoculation). d, Therapeutic antitumour activity against B16F10 tumours in mice (B16‐M27, Trp2 n = 8; B16‐M30 n = 7; others n = 10) conferred by immunization with epitopes encoding immunogenic B16F10 mutations or an immunodominant wild type Trp2 epitope6. The area under the tumour growth curve at day 30 after tumour inoculation was normalized to untreated control mice and depicted as mean ± s.e.m. Red and black columns represent mutations recognized by CD8+ or CD4+ T cells, respectively. e, Spontaneous immune responses in splenocytes of irrelevant RNA treated B16F10 tumour bearing C57BL/6 mice (n = 3) were tested by ELISpot for recognition of peptides (mean + s.e.m.).
Extended Data Figure 3 Mechanism of antitumour activity of mutation specific poly‐epitope vaccines in CT26 tumour‐bearing mice.
a, BALB/c mice (n = 5) were vaccinated either with pentatope (35 µg) or the corresponding mixture of five RNA monotopes (7 µg each). T‐cell responses in peptide‐stimulated splenocytes of mice were measured ex vivo via ELISpot (medium control subtracted mean ± s.e.m.). b, c, BALB/c mice (n = 10) were inoculated i.v. with CT26 tumour cells and left untreated or injected with irrelevant, CT26‐M19 or pentatope1 or 2 RNA in absence (b) or presence of a CD8 T cell depleting antibody or a CD40L blocking antibody (c). Mean ± s.e.m. of tumour nodules per lung are shown. d, Immunofluorescence analyses of tumour‐infiltrating lymphocytes in pentatope2‐vaccinated mice. Upper panel, lung tumour tissue stained for CD4 and CD8 or CD4 and FoxP3. Scale bar, 50 µm. Lower panel left, proportion of infiltrating cells in sections of irrelevant (CD4: n = 13; CD8 = 9; FoxP3: n = 13) or pentatope (CD4: n = 17; CD8: n = 6; FoxP3: n = 10) RNA‐treated animals. Lower panel right, tumour area in sections of control (n = 22) and pentatope2‐treated (n = 20) animals (mean ± s.e.m.).
Extended Data Figure 4 Immunogenicity testing of PME pentatope‐encoded mutations.
Splenocytes of PME RNA vaccinated BALB/c mice (n = 6) were tested ex vivo for recognition of peptides representing the mutated 27mer sequences represented in PME pentatopes with or without addition of an MHC class II‐blocking antibody. Mean + s.e.m. of background (medium control) subtracted responses are shown.
Extended Data Table 1 Immunogenic B16F10 mutations
Extended Data Table 2 Immunogenic CT26 mutations
Extended Data Table 3 Immunogenic 4T1 mutations
Extended Data Table 4 CT26 mutated epitopes encoded in pentatope 1+2
Extended Data Table 5 In silico prediction of CT26 mutations with abundant expression and favourable MHC class II binding properties
PowerPoint slides
Rights and permissions
About this article
Cite this article
Kreiter, S., Vormehr, M., van de Roemer, N. et al. Mutant MHC class II epitopes drive therapeutic immune responses to cancer.Nature 520, 692–696 (2015). https://doi.org/10.1038/nature14426
- Received: 07 November 2014
- Accepted: 30 March 2015
- Published: 22 April 2015
- Issue Date: 30 April 2015
- DOI: https://doi.org/10.1038/nature14426