Skip to main content
Log in

Tumorvakzinierung –Strategien und Timing

Tumor vaccination—strategies and time points

  • Arzneimitteltherapie
  • Published:
Der Internist Aims and scope Submit manuscript

Zusammenfassung

Hintergrund

Immuntherapien haben in den vergangenen Jahren einen zunehmenden Stellenwert in der Behandlung maligner Erkrankungen gewonnen. Hierzu gehören auch Tumorvakzine, die therapeutisch eingesetzt werden, um das Immunsystem spezifisch gegen Tumorzellen zu richten.

Fragestellung

Es werden verschiedene Strategien der Tumorvakzinierungen, ihr aktueller Entwicklungstand, der optimale Einsatzzeitpunkt und Kombinationsmöglichkeiten in der Therapie von malignen Erkrankungen diskutiert.

Methoden

Wissenschaftliche Publikationen zu unterschiedlichen Tumorvakzinierungsstrategien unter Einbeziehung aktueller Studien werden zusammengefasst.

Schlussfolgerung

Für eine effektive Tumorvakzinierung ist die Auswahl geeigneter Tumorantigene, die über Moleküle der humanen Leukozytenantigene (HLA) auf der Zelloberfläche präsentiert werden, essenziell. Geeignete Antigene müssen sowohl exklusiv auf den Tumorzellen präsentiert werden als auch eine spezifische Antitumorimmunantwort induzieren, d. h. zytotoxische und T‑Helferzellen aktivieren. Hierfür kommen einerseits Neoepitope, die aus tumorspezifischen Mutationen stammen, und andererseits tumorassoziierte Antigene (TAA), die durch veränderte Genexpression oder Prozessierung ausschließlich im Tumorgewebe präsentiert sind, infrage. Für die Applikation der Antigene stehen verschiedenste Strategien, wie u. a. Peptidvakzine, DNS- oder RNS-basierte Vakzine sowie auf dendritischen Zellen oder kompletten Tumorzellen basierende Ansätze, kombiniert mit geeigneten Adjuvanzien zur Verfügung. Derzeit wird die Wirksamkeit zahlreicher Vakzinierungskonzepte und Kombinationsprotokolle in klinischen Studien im Hinblick auf Tumorreduktion und Rezidivprävention evaluiert.

Abstract

Background

Immunotherapies have gained increasing importance in the treatment of cancer in recent years. This also includes tumor vaccines, which are used therapeutically to direct the immune system specifically against tumor cells.

Objectives

Different strategies of tumor vaccination, their current state of development, the optimal timing and possible combinations of cancer vaccines in the treatment of cancer are discussed.

Methods

Scientific publications on various tumor vaccination strategies based on ongoing studies that are listed on clinicaltrials.gov are summarized.

Conclusions

For effective tumor vaccination, the selection of suitable tumor antigens present on the cell surface via human leukocyte antigen (HLA) molecules is essential. Suitable antigens should be present exclusively on tumor cells and able to induce a specific anti-tumor immune response, i.e. activate cytotoxic and T helper cells. For this purpose, neoepitopes derived from tumor-specific mutations or tumor-associated antigens (TAAs), which are present exclusively in tumor tissue due to altered gene expression or processing, can be used. For the application of the antigens, various strategies combined with suitable adjuvants are available, including peptide vaccines, DNA- or RNA-based vaccines, approaches with dendritic cells or whole tumor cell vaccines. Currently, numerous vaccination approaches as well as combination protocols are being evaluated in clinical trials with the aim to establish specific and low side effect immunotherapies to combat malignancies and enable long-term protection from disease recurrence via the induction of long-lasting antitumor immune responses.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Abb. 1
Abb. 2

Literatur

  1. Antonilli M, Rahimi H, Visconti V et al (2016) Triple peptide vaccination as consolidation treatment in women affected by ovarian and breast cancer: clinical and immunological data of a phase I/II clinical trial. Int J Oncol 48(4):1369–1378. https://doi.org/10.3892/ijo.2016.3386

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Bilich T, Nelde A, Bichmann L et al (2019) The HLA ligandome landscape of chronic myeloid leukemia delineates novel T‑cell epitopes for immunotherapy. Blood 133:550–565

    Article  CAS  Google Scholar 

  3. Brocks D, Schmidt CR, Daskalakis M et al (2017) DNMT and HDAC inhibitors induce cryptic transcription start sites encoded in long terminal repeats. Nat Genet 49:1052–1060

    Article  CAS  Google Scholar 

  4. Coukos G (2020) Neoadjuvant immune-checkpoint blockade in resectable colon cancer. Nat Med 26:473–474

    Article  CAS  Google Scholar 

  5. Federico P, Petrillo A, Giordano P et al (2020) Immune checkpoint inhibitors in hepatocellular carcinoma: current status and novel perspectives. Cancers 12(10):3025. https://doi.org/10.3390/cancers12103025

    Article  CAS  PubMed Central  Google Scholar 

  6. Haen SP, Loffler MW, Rammensee HG et al (2020) Towards new horizons: characterization, classification and implications of the tumour antigenic repertoire. Nat Rev Clin Oncol 17:595–610

    Article  Google Scholar 

  7. Harzstark AL, Small EJ (2007) Immunotherapy for prostate cancer using antigen-loaded antigen-presenting cells: APC8015 (Provenge). Expert Opin Biol Ther 7:1275–1280

    Article  CAS  Google Scholar 

  8. Hilf N, Kuttruff-Coqui S, Frenzel K et al (2019) Actively personalized vaccination trial for newly diagnosed glioblastoma. Nature 565:240–245

    Article  CAS  Google Scholar 

  9. Hollingsworth RE, Jansen K (2019) Turning the corner on therapeutic cancer vaccines. NPJ Vaccines 4:7

    Article  Google Scholar 

  10. Iversen TZ, Engell-Noerregaard L, Ellebaek E et al (2014) Long-lasting disease stabilization in the absence of toxicity in metastatic lung cancer patients vaccinated with an epitope derived from indoleamine 2,3 dioxygenase. Clin Cancer Res 20:221–232

    Article  CAS  Google Scholar 

  11. Khong H, Overwijk WW (2016) Adjuvants for peptide-based cancer vaccines. J Immunother Cancer 4:56

    Article  Google Scholar 

  12. Kowalewski DJ, Schuster H, Backert L et al (2015) HLA ligandome analysis identifies the underlying specificities of spontaneous antileukemia immune responses in chronic lymphocytic leukemia (CLL). Proc Natl Acad Sci U S A 112:E166–E175

    Article  CAS  Google Scholar 

  13. Kowalewski DJ, Walz S, Backert L et al (2016) Carfilzomib alters the HLA-presented peptidome of myeloma cells and impairs presentation of peptides with aromatic C‑termini. Blood Cancer J 6:e411

    Article  CAS  Google Scholar 

  14. Kreiter S, Vormehr M, Van De Roemer N et al (2015) Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature 520:692–696

    Article  CAS  Google Scholar 

  15. Laheru D, Lutz E, Burke J et al (2008) Allogeneic granulocyte macrophage colony-stimulating factor-secreting tumor immunotherapy alone or in sequence with cyclophosphamide for metastatic pancreatic cancer: a pilot study of safety, feasibility, and immune activation. Clin Cancer Res 14:1455–1463

    Article  CAS  Google Scholar 

  16. Le DT, Lutz E, Uram JN et al (2013) Evaluation of ipilimumab in combination with allogeneic pancreatic tumor cells transfected with a GM-CSF gene in previously treated pancreatic cancer. J Immunother 36:382–389

    Article  CAS  Google Scholar 

  17. Mcnamara MA, Nair SK, Holl EK (2015) RNA-based vaccines in cancer immunotherapy. J Immunol Res 2015:794528

    Article  Google Scholar 

  18. Mennonna D, Maccalli C, Romano MC et al (2017) T cell neoepitope discovery in colorectal cancer by high throughput profiling of somatic mutations in expressed genes. Gut 66:454–463

    Article  CAS  Google Scholar 

  19. Murphy K, Travers P, Walport M et al (2012) Janeway’s immunobiology. Garland Science, New York

    Google Scholar 

  20. Nelde A, Rammensee HG, Walz JS (2021) The peptide vaccine of the future. Mol Cell Proteomics 20:100022

    Article  CAS  Google Scholar 

  21. Ott PA, Hu Z, Keskin DB et al (2017) An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 547:217–221

    Article  CAS  Google Scholar 

  22. Overwijk WW (2017) Cancer vaccines in the era of checkpoint blockade: the magic is in the adjuvant. Curr Opin Immunol 47:103–109

    Article  CAS  Google Scholar 

  23. Pardoll DM (2012) The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 12:252–264

    Article  CAS  Google Scholar 

  24. Peoples GE, Gurney JM, Hueman MT et al (2005) Clinical trial results of a HER2/neu (E75) vaccine to prevent recurrence in high-risk breast cancer patients. J Clin Oncol 23:7536–7545

    Article  CAS  Google Scholar 

  25. Petrini B, Wasserman J, Glas U et al (1982) T lymphocyte subpopulations in blood following radiation therapy for breast cancer. Eur J Cancer Clin Oncol 18:921–924

    Article  CAS  Google Scholar 

  26. Queirolo P, Boutros A, Tanda E et al (2019) Immune-checkpoint inhibitors for the treatment of metastatic melanoma: a model of cancer immunotherapy. Semin Cancer Biol 59:290–297

    Article  CAS  Google Scholar 

  27. Sabado RL, Balan S, Bhardwaj N (2017) Dendritic cell-based immunotherapy. Cell Res 27:74–95

    Article  CAS  Google Scholar 

  28. Sahin U, Oehm P, Derhovanessian E et al (2020) An RNA vaccine drives immunity in checkpoint-inhibitor-treated melanoma. Nature 585:107–112

    Article  CAS  Google Scholar 

  29. Smith BD, Kasamon YL, Kowalski J et al (2010) K562/GM-CSF immunotherapy reduces tumor burden in chronic myeloid leukemia patients with residual disease on imatinib mesylate. Clin Cancer Res 16:338–347

    Article  CAS  Google Scholar 

  30. Srivatsan S, Patel JM, Bozeman EN et al (2014) Allogeneic tumor cell vaccines: the promise and limitations in clinical trials. Hum Vaccin Immunother 10:52–63

    Article  CAS  Google Scholar 

  31. Thomas S, Prendergast GC (2016) Cancer vaccines: a brief overview. Methods Mol Biol 1403:755–761

    Article  Google Scholar 

  32. Weihrauch MR, Ansen S, Jurkiewicz E et al (2005) Phase I/II combined chemoimmunotherapy with carcinoembryonic antigen-derived HLA-A2-restricted CAP‑1 peptide and irinotecan, 5‑fluorouracil, and leucovorin in patients with primary metastatic colorectal cancer. Clin Cancer Res 11:5993–6001

    Article  CAS  Google Scholar 

  33. Westermann J, Florcken A, Willimsky G et al (2011) Allogeneic gene-modified tumor cells (RCC-26/IL-7/CD80) as a vaccine in patients with metastatic renal cell cancer: a clinical phase‑I study. Gene Ther 18:354–363

    Article  CAS  Google Scholar 

  34. Zitvogel L, Apetoh L, Ghiringhelli F et al (2008) Immunological aspects of cancer chemotherapy. Nat Rev Immunol 8:59–73

    Article  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Juliane S. Walz.

Ethics declarations

Interessenkonflikt

Y. Maringer und J.S. Walz geben an, dass kein Interessenkonflikt besteht.

Für diesen Beitrag wurden von den Autoren keine Studien an Menschen oder Tieren durchgeführt. Für die aufgeführten Studien gelten die jeweils dort angegebenen ethischen Richtlinien.

Additional information

Redaktion

M. Wehling, Mannheim

Erstveröffentlichung in Der Gastroenterologe (2021) 16:241–248. https://doi.org/10.1007/s11377-021-00532-4

figure qr

QR-Code scannen & Beitrag online lesen

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Maringer, Y., Walz, J.S. Tumorvakzinierung –Strategien und Timing. Internist 62, 991–997 (2021). https://doi.org/10.1007/s00108-021-01138-2

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00108-021-01138-2

Schlüsselwörter

Keywords

Navigation