Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer

Abstract

T cells directed against mutant neo-epitopes drive cancer immunity. However, spontaneous immune recognition of mutations is inefficient. We recently introduced the concept of individualized mutanome vaccines and implemented an RNA-based poly-neo-epitope approach to mobilize immunity against a spectrum of cancer mutations1,2. Here we report the first-in-human application of this concept in melanoma. We set up a process comprising comprehensive identification of individual mutations, computational prediction of neo-epitopes, and design and manufacturing of a vaccine unique for each patient. All patients developed T cell responses against multiple vaccine neo-epitopes at up to high single-digit percentages. Vaccine-induced T cell infiltration and neo-epitope-specific killing of autologous tumour cells were shown in post-vaccination resected metastases from two patients. The cumulative rate of metastatic events was highly significantly reduced after the start of vaccination, resulting in a sustained progression-free survival. Two of the five patients with metastatic disease experienced vaccine-related objective responses. One of these patients had a late relapse owing to outgrowth of β2-microglobulin-deficient melanoma cells as an acquired resistance mechanism. A third patient developed a complete response to vaccination in combination with PD-1 blockade therapy. Our study demonstrates that individual mutations can be exploited, thereby opening a path to personalized immunotherapy for patients with cancer.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Broad mobilization of mutation-specific immunity by vaccination.
Figure 2: Rapid expansion of neo-epitope-specific T cells with central and effector memory phenotypes by vaccination.
Figure 3: Disease control by vaccination in melanoma patients with high risk of relapse.
Figure 4: Neo-epitope-induced CTL responses associated with immune escape by outgrowth of B2M-deficient melanoma cells in P04.

Similar content being viewed by others

References

  1. Castle, J. C. et al. Exploiting the mutanome for tumor vaccination. Cancer Res. 72, 1081–1091 (2012)

    Article  CAS  PubMed  Google Scholar 

  2. Kreiter, S. et al. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature 520, 692–696 (2015)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  3. 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  PubMed  PubMed Central  Google Scholar 

  4. Snyder, A. et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 371, 2189–2199 (2014)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Van Allen, E. M. et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 350, 207–211 (2015)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  6. McGranahan, N. et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 351, 1463–1469 (2016)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Tran, E. et al. T-cell transfer therapy targeting mutant KRAS in cancer. N. Engl. J. Med. 375, 2255–2262 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Linnemann, C. et al. High-throughput epitope discovery reveals frequent recognition of neo-antigens by CD4+ T cells in human melanoma. Nat. Med. 21, 81–85 (2015)

    Article  CAS  PubMed  Google Scholar 

  9. Matsushita, H. et al. Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting. Nature 482, 400–404 (2012)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  10. Tran, E. et al. Immunogenicity of somatic mutations in human gastrointestinal cancers. Science 350, 1387–1390 (2015)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  11. Yadav, M. et al. Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing. Nature 515, 572–576 (2014)

    Article  ADS  CAS  PubMed  Google Scholar 

  12. Gubin, M. M. et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 515, 577–581 (2014)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  13. Carreno, B. M. et al. Cancer immunotherapy. A dendritic cell vaccine increases the breadth and diversity of melanoma neoantigen-specific T cells. Science 348, 803–808 (2015)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  14. Delamarre, L., Mellman, I. & Yadav, M. Cancer immunotherapy. Neo approaches to cancer vaccines. Science 348, 760–761 (2015)

    CAS  PubMed  Google Scholar 

  15. 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  PubMed  Google Scholar 

  16. Bassani-Sternberg, M. et al. Direct identification of clinically relevant neoepitopes presented on native human melanoma tissue by mass spectrometry. Nat. Commun. 7, 13404 (2016)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. Abelin, J. G. et al. Mass spectrometry profiling of HLA-associated peptidomes in mono-allelic cells enables more accurate epitope prediction. Immunity 46, 315–326 (2017)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Chen, D. S. & Mellman, I. Elements of cancer immunity and the cancer-immune set point. Nature 541, 321–330 (2017)

    Article  ADS  CAS  PubMed  Google Scholar 

  19. Tumeh, P. C. et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515, 568–571 (2014)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Robert, C. et al. Pembrolizumab versus ipilimumab in advanced melanoma. N. Engl. J. Med. 372, 2521–2532 (2015)

    Article  CAS  PubMed  Google Scholar 

  21. D’Urso, C. M. et al. Lack of HLA class I antigen expression by cultured melanoma cells FO-1 due to a defect in B2m gene expression. J. Clin. Invest. 87, 284–292 (1991)

    Article  PubMed  PubMed Central  Google Scholar 

  22. Zaretsky, J. M. et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N. Engl. J. Med. 375, 819–829 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Melero, I. et al. Evolving synergistic combinations of targeted immunotherapies to combat cancer. Nat. Rev. Cancer 15, 457–472 (2015)

    Article  CAS  PubMed  Google Scholar 

  24. Wolchok, J. D. et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin. Cancer Res. 15, 7412–7420 (2009)

    Article  CAS  PubMed  Google Scholar 

  25. Dudley, M. E., Wunderlich, J. R., Shelton, T. E., Even, J. & Rosenberg, S. A. Generation of tumor-infiltrating lymphocyte cultures for use in adoptive transfer therapy for melanoma patients. J. Immunother. 26, 332–342 (2003)

    Article  PubMed  PubMed Central  Google Scholar 

  26. 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  PubMed  Google Scholar 

  27. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Mortazavi, A., Williams, B. A., McCue, K., Schaeffer, L. & Wold, B. Mapping and quantifying mammalian transcriptomes by RNA-seq. Nat. Methods 5, 621–628 (2008)

    Article  CAS  PubMed  Google Scholar 

  30. Kim, Y. et al. Immune epitope database analysis resource. Nucleic Acids Res. 40, W525–W530 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Untergasser, A. et al. Primer3—new capabilities and interfaces. Nucleic Acids Res. 40, e115 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Koressaar, T. & Remm, M. Enhancements and modifications of primer design program Primer3. Bioinformatics 23, 1289–1291 (2007)

    Article  CAS  PubMed  Google Scholar 

  33. Kent, W. J. BLAT—the BLAST-like alignment tool. Genome Res. 12, 656–664 (2002)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. 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  PubMed  Google Scholar 

  35. Grudzien-Nogalska, E. et al. Synthetic mRNAs with superior translation and stability properties. Methods Mol. Biol. 969, 55–72 (2013)

    Article  CAS  PubMed  Google Scholar 

  36. 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  PubMed  Google Scholar 

  37. Berensmeier, S. Magnetic particles for the separation and purification of nucleic acids. Appl. Microbiol. Biotechnol. 73, 495–504 (2006)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Simon, P. et al. Functional TCR retrieval from single antigen-specific human T cells reveals multiple novel epitopes. Cancer Immunol. Res. 2, 1230–1244 (2014)

    CAS  Google Scholar 

  39. Brochet, X., Lefranc, M.-P. & Giudicelli, V. IMGT/V-QUEST: the highly customized and integrated system for IG and TR standardized V-J and V-D-J sequence analysis. Nucleic Acids Res. 36, W503–W508 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Thorvaldsdóttir, H., Robinson, J. T. & Mesirov, J. P. Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief. Bioinform. 14, 178–192 (2013)

    Article  PubMed  CAS  Google Scholar 

  41. Zhao, Q.-Y. et al. Optimizing de novo transcriptome assembly from short-read RNA-Seq data: a comparative study. BMC Bioinformatics 12 (Suppl 14), S2 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Omokoko, T. A. et al. Luciferase mRNA transfection of antigen presenting cells permits sensitive nonradioactive measurement of cellular and humoral cytotoxicity. J. Immunol. Res. 2016, 9540975 (2016)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank J. de Graaf, I. Eichelbrönner, L. Leppin, L. Giese and S. Vogler, D. Becker, M. Dorner, J. Grützner, M. Hossainzadeh, A. Selmi, S. Wessel, C. Ecker, M. Lochschmitt, B. Schmitz, C. Anft, N. Bidmon, H. Schröder, D. Barea Roldán, C. Walter, S. Wöll, C. Rohde, O. Renz, F. Bayer, C. Kröner, B. Otte, T. Stricker, M. Drude S. Petri, M. Mechler, L. Hebich, B. Steege, A. Oelbermann, J. Schwarz, C. Britten, J. C. Castle and B. Pless for technical support, project management and advice. We thank A. Tüttenberg for support with a figure. We thank I. Mellman, L. Delamarre and G. Fine for critical reading of the manuscript. We thank K. Sahin for her advice. The study was supported by the CI3 cluster program of the Federal Ministry of Education and Research (BMBF).

Author information

Authors and Affiliations

Authors

Contributions

U.S. conceptualized the work and strategy. E.D., Pe.Si., T.O., I.O., I.V., S.A., A.R. and B.K. planned and analysed experiments. E.G., R.R., A.B., C.T. and A.H. did experiments. M.L., V.B., A.D.T., B.S., C.A., A.P. and Pa.So. performed and analysed NGS runs. A.N.K., J.B. and J.C. manufactured the RNA vaccines, O.W., M.W. and M.Z. performed quality assurance. M.M., B.K., S.H., K.H.S., F.M., A.K.-B., D.L. and S.B. managed sample logistics. R.N., C.G., S.G., C.H., J.U. are clinical investigators. C.L. is the principal clinical investigator. U.L., J.D., M.D. and S.K. supported clinical grade assays. U.S., ÖT supported by E.D., M.V., Pe.Si., M.L, M.M., B.S. interpreted data and wrote the manuscript. All authors supported the revision of the manuscript.

Corresponding author

Correspondence to Ugur Sahin.

Ethics declarations

Competing interests

Some of the authors are employees at BioNTech AG (Mainz, Germany) as mentioned in the affiliations. U.S. is stock owner of BioNTech AG (Mainz, Germany). U.S., M.L., B.S., M.V., A.N.K., M.D., A.T., Ö.T. and S.K. are inventors on patents and patent applications, which cover parts of this article.

Additional information

Reviewer Information Nature thanks C. Melief and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Publisher's note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Figure 1 Expressed non-synonymous mutations in patient melanomas are mostly C>T and G>A transitions.

Extended Data Figure 2 Characterization of and examples for CD4+CD8+ and wild-type cross-reactive T cell responses induced by neo-epitope vaccination.

a, Pre- and post-vaccination T cell frequencies against neo-epitope-encoding RNAs (nine patients). In vitro stimulation of T cells as in Fig. 1f, except autologous DCs for read-out were loaded with RNA encoding single neo-epitopes instead of OLPs. 88% (110 out of 125) concordance rate of both measurements, with slightly higher sensitivity of the OLP read-out. b, c, Pre- and post-vaccination CD4+ and CD8+ T-cell-enriched cultures of patient P19 stimulated with the patient’s pentatope RNAs read-out against autologous DCs loaded with OLPs representing mutated neo-epitopes in the ST5 and UTP6 proteins. b, Example for an exclusive de novo CD4+ T cell response. c, Example for a de novo response recognized by both CD4+ and CD8+ T cells at different portions of the mutated sequence. d, CD4+ and CD8+ T cell cultures were quality controlled for purity after stimulation by flow cytometry to exclude cross contamination of the separated in vitro stimulation cultures. e, Wild-type epitope cross-reactivity of post-vaccine CD4+ (top left) and CD8+ (top right) T cells (tested for 55 neo-epitopes in total). Autologous DCs loaded with RNA (not marked), with different OLPs (P1, P2) of the OLP pool encoding the mutated or the wild-type sequence used as read out (marked with ‘P’). Examples of reactivity with mutated and wild-type epitopes analysed by ELISpot is shown for P04 CDC37L1(P186L). fh, Three examples of wild-type epitope cross-reactive responses. For all three responses immune recognition of autologous DCs that naturally express the wild-type genes was not detected. g, Sanger sequencing of P05 FAM135B in control DCs used in ELISpot (f). Control RNA, luciferase. h, Left, example for wild-type reactivity observed with encoding RNA only but not with OLPs. Right, differential cross-reactivity depended on the assessed OLP in a single T cell response.

Extended Data Figure 3 Characterization of selected TCRs obtained by single-cell cloning.

a, Specificity of NARFL(E62K)-specific TCRs cloned from CD8+ T cells of patient P01. CD8+ T cells transfected with four TCRs directed against a mutation in the NARFL gene were tested by ELISpot for recognition of HLA-A*3101-transfected K562 cells pulsed with individual 15mer OLPs representing the mutant or the wild-type sequence. Control, irrelevant OLPs. bg, Cloning and characterization of TCRs directed against mutations in PPFIA4 and HPN proteins of P02. b, e, Activation-induced IFNγ-secretion-based single-cell sorting from in vitro stimulation cultures of neo-epitope-specific T cells after co-culture with autologous DCs transfected with RNA (b) or pulsed with OLPs (e) encoding the respective neo-epitope. Control, RNA or OLPs encoding an irrelevant neo-epitope. c, d, f, g, Determination of HLA-restriction and specificity of the cloned TCRs by ELISpot with healthy donor-derived T cells co-transfected with RNAs encoding the identified TCR-α/β chains and peptide-pulsed K562 cells expressing single HLA molecules of the patient. Controls, OLPs encoding an irrelevant neo-epitope (c), HIV-gag (f), target cells without OLPs (d, g), Staphylococcal-enterotoxin-B (SEB) (g).

Extended Data Figure 4 Kinetics and specificity of selected vaccine-induced T cell responses.

a, Kinetics of vaccine-induced responses ex vivo by ELISpot (first three) or multimer staining (last two). b, Reactivity of CD8+ T cells of patient P05 against autologous DCs loaded with individual OLPs or with the predicted HLA-B*0702-restricted minimal epitope (ME, bold sequence). c, Detection of CD8+ T cells recognizing minimal epitope in the same in vitro stimulation cultures by HLA multimer staining. df, CD8+ T cell responses of patient P17 against two different HLA-restricted T cell epitopes generated by the same mutation. d, Detection of CD8+ T cells recognizing HSCVMASLR, the best-predicted HLA-A*6801-restricted minimal epitope within P17 RETSAT(P546) (contained in OLP 3 and 4) in post-vaccination TILs from patient P17 by multimer staining. e, ELISpot assay of post-vaccine CD8+ T cells of P17 on autologous DCs loaded with individual neo-epitope OLPs. f, Specificity of two HLA-B*3701-restricted RETSAT(P546S) TCRs obtained from TILs of patient P17 recognizing OLP 1 and 2. Control, HIV-gag OLPs. g, Reactivity of three unvaccinated healthy blood bank donors with predicted neo-epitopes found to be strongly recognized by patients (four neo-epitopes from two patients) by multimer staining.

Extended Data Figure 5 Characterization of vaccine-associated immune responses, tumour immune infiltrates and B2M loss in P04.

a, Time-course analysis of selected T cell responses in blood PBMCs by multimers. b, c, TCRs cloned from neo-epitope-specific CD8+ T cells transfected into healthy donor T cells analysed by ELISpot assay for recognition of K562-A*0201 cells transfected with RNAs encoding the neo-epitopes (b) and loaded with minimal epitopes (c) (FLNA(P639L), HIAKSLFEV; CDC37L1(P183L), FLSDHLYLV). Triplicates (mean ± s.d.) are shown. Controls, respective other minimal peptide epitope. df, Comparison of pre-vaccination (lymph node metastasis inguinal right; used for vaccine neo-epitope selection) and post-vaccination (lymph node metastasis iliacal right). d, T cell infiltrates analysed by immunohistochemistry. Tumour cells visualized by anti-Melan-A co-staining. e, Differential gene expression patterns comparing pre- and post-vaccination tumour samples. f, Complete B2M locus deletion in MZ-GaBa-018 but not in the pre-vaccination tumour resectate and autologous PBMCs as shown in coverage profiles of exome data. Deletion start localized in an intron in the PATL2 locus upstream of B2M in the exome capture. g, Killing of MZ-GaBa-018 melanoma cells by neo-epitope RNA-stimulated T cells. Untreated, IFNγ pre-treated or B2M-RNA-transfected melanoma cells used as targets for autologous CD8+ or CD4+ T cells from stimulation cultures (effector:target ratio 50:1). Effects on HLA class I/II surface staining analysed by flow cytometry (lower panel). h, Neo-epitope RNA transcript levels expressed by the autologous tumour cell line MZ-GaBa-018 as analysed from RNA-seq data and plotted as RPKM values.

Extended Data Table 1 Patient baseline characteristics
Extended Data Table 2 Overview of immune responses in all 13 patients
Extended Data Table 3 Identified minimal HLA-class-I-restricted T cell epitopes
Extended Data Table 4 Neo-epitope-specific TCR-α/β chains cloned from single T cells of four melanoma patients
Extended Data Table 5 Patient characteristics and clinical events

Supplementary information

Supplementary Table 1

This table lists the neo-epitope vaccine sequence for patient P04. (XLSX 17 kb)

Supplementary Table 2

This table summarizes all identified neo-epitopes across patients. (XLSX 36 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sahin, U., Derhovanessian, E., Miller, M. et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 547, 222–226 (2017). https://doi.org/10.1038/nature23003

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature23003

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research