A novel anti-HER2 anthracycline-based antibody-drug conjugate induces adaptive anti-tumor immunity and potentiates PD-1 blockade in breast cancer
Jazyk angličtina Země Anglie, Velká Británie Médium electronic
Typ dokumentu časopisecké články, práce podpořená grantem
PubMed
30665463
PubMed Central
PMC6341578
DOI
10.1186/s40425-018-0464-1
PII: 10.1186/s40425-018-0464-1
Knihovny.cz E-zdroje
- Klíčová slova
- Anthracycline, Antibody-drug conjugates, Checkpoint inhibitor combination therapy, HER2-positive breast cancer,
- MeSH
- antigeny CD279 antagonisté a inhibitory MeSH
- antracykliny terapeutické užití MeSH
- CD8-pozitivní T-lymfocyty účinky léků imunologie MeSH
- experimentální nádory mléčných žláz farmakoterapie imunologie MeSH
- imunokonjugáty terapeutické užití MeSH
- imunologická paměť účinky léků MeSH
- lidé MeSH
- myši inbrední BALB C MeSH
- nádorové buněčné linie MeSH
- protinádorové látky imunologicky aktivní terapeutické užití MeSH
- receptor erbB-2 antagonisté a inhibitory genetika MeSH
- trastuzumab terapeutické užití MeSH
- zvířata MeSH
- Check Tag
- lidé MeSH
- ženské pohlaví MeSH
- zvířata MeSH
- Publikační typ
- časopisecké články MeSH
- práce podpořená grantem MeSH
- Názvy látek
- antigeny CD279 MeSH
- antracykliny MeSH
- ERBB2 protein, human MeSH Prohlížeč
- imunokonjugáty MeSH
- Pdcd1 protein, mouse MeSH Prohlížeč
- protinádorové látky imunologicky aktivní MeSH
- receptor erbB-2 MeSH
- trastuzumab MeSH
Increasing evidence suggests that antibody-drug conjugates (ADCs) can enhance anti-tumor immunity and improve clinical outcome. Here, we elucidate the therapeutic efficacy and immune-mediated mechanisms of a novel HER2-targeting ADC bearing a potent anthracycline derivate as payload (T-PNU) in a human HER2-expressing syngeneic breast cancer model resistant to trastuzumab and ado-trastuzumab emtansine. Mechanistically, the anthracycline component of the novel ADC induced immunogenic cell death leading to exposure and secretion of danger-associated molecular signals. RNA sequencing derived immunogenomic signatures and TCRβ clonotype analysis of tumor-infiltrating lymphocytes revealed a prominent role of the adaptive immune system in the regulation of T-PNU mediated anti-cancer activity. Depletion of CD8 T cells severely reduced T-PNU efficacy, thus confirming the role of cytotoxic T cells as drivers of the T-PNU mediated anti-tumor immune response. Furthermore, T-PNU therapy promoted immunological memory formation in tumor-bearing animals protecting those from tumor rechallenge. Finally, the combination of T-PNU and checkpoint inhibition, such as α-PD1, significantly enhanced tumor eradication following the treatment. In summary, a novel PNU-armed, HER2-targeting ADC elicited long-lasting immune protection in a murine orthotopic breast cancer model resistant to other HER2-directed therapies. Our findings delineate the therapeutic potential of this novel ADC payload and support its clinical development for breast cancer patients and potentially other HER2 expressing malignancies.
Cancer Immunology Department of Biomedicine University of Basel Basel Switzerland
Celonic AG Eulerstrasse 55 4051 Basel Switzerland
Department of Biosystems Science and Engineering ETH Zürich Basel Switzerland
Institute of Pathology University Hospital Basel Basel Switzerland
NBE Therapeutics Ltd Hochbergerstrasse 60C 4057 Basel Switzerland
Zobrazit více v PubMed
Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science. 1987;235:177–182. doi: 10.1126/science.3798106. PubMed DOI
Dawood S, Broglio K, Buzdar AU, Hortobagyi GN, Giordano SH. Prognosis of women with metastatic breast cancer by HER2 status and trastuzumab treatment: an institutional-based review. J Clin Oncol Proc Am Soc Clin Oncol. 2010;28:92–98. doi: 10.1200/JCO.2008.19.9844. PubMed DOI PMC
Hudis CA. Trastuzumab--mechanism of action and use in clinical practice. N Engl J Med. 2007;357:39–51. doi: 10.1056/NEJMra043186. PubMed DOI
Lewis Phillips GD, Li G, Dugger DL, Crocker LM, Parsons KL, Mai E, et al. Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res. American Association for Cancer Research. 2008;68:9280–9290. PubMed
Krop IE, Kim S-B, González-Martín A, LoRusso PM, Ferrero J-M, Smitt M, et al. Trastuzumab emtansine versus treatment of physician's choice for pretreated HER2-positive advanced breast cancer (TH3RESA): a randomised, open-label, phase 3 trial. Lancet Oncol. 2014;15:689–699. doi: 10.1016/S1470-2045(14)70178-0. PubMed DOI
Verma S, Miles D, Gianni L, Krop IE, Welslau M, Baselga J, et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med Massachusetts Medical Society. 2012;367:1783–1791. doi: 10.1056/NEJMoa1209124. PubMed DOI PMC
Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, adaptive, and acquired resistance to Cancer immunotherapy. Cell. 2017;168:707–723. doi: 10.1016/j.cell.2017.01.017. PubMed DOI PMC
Gerber H-P, Sapra P, Loganzo F, May C. Combining antibody-drug conjugates and immune-mediated cancer therapy: what to expect? Biochem Pharmacol. 2016;102:1–6. doi: 10.1016/j.bcp.2015.12.008. PubMed DOI
Müller P, Kreuzaler M, Khan T, Thommen DS, Martin K, Glatz K, et al. Trastuzumab emtansine (T-DM1) renders HER2+ breast cancer highly susceptible to CTLA-4/PD-1 blockade. Sci Transl Med. American Association for the Advancement of Science; 2015;7:315ra188–8. PubMed
Polakis P. Antibody Drug Conjugates for Cancer Therapy. Esbenshade TA, editor. Pharmacol Rev American Society for Pharmacology and Experimental Therapeutics; 2016;68:3–19. PubMed
Beerli RR, Hell T, Merkel AS, Grawunder U. Sortase Enzyme-Mediated Generation of Site-Specifically Conjugated Antibody Drug Conjugates with High In Vitro and In Vivo Potency. Hagemeyer CE, editor. PLoS ONE. 2015;10:e0131177. PubMed PMC
Stefan N, Gébleux R, Waldmeier L, Hell T, Escher M, Wolter FI, et al. Highly Potent, Anthracycline-based Antibody-Drug Conjugates Generated by Enzymatic, Site-specific Conjugation. Mol Cancer Ther American Association for Cancer Research. 2017;16:879–892. doi: 10.1158/1535-7163.MCT-16-0688. PubMed DOI
Yu S-F, Zheng B, Go M, Lau J, Spencer S, Raab H, et al. A Novel Anti-CD22 Anthracycline-Based Antibody-Drug Conjugate (ADC) That Overcomes Resistance to Auristatin-Based ADCs. Clin Cancer Res American Association for Cancer Research. 2015;21:3298–3306. doi: 10.1158/1078-0432.CCR-14-2035. PubMed DOI
Junttila MR, Mao W, Wang X, Wang B-E, Pham T, Flygare J, et al. Targeting LGR5+ cells with an antibody-drug conjugate for the treatment of colon cancer. Sci Transl Med American Association for the Advancement of Science; 2015;7:314ra186–6. PubMed
Orsini F, Pavelic Z, Mihich E. Increased primary cell-mediated immunity in culture subsequent to adriamycin or daunorubicin treatment of spleen donor mice. Cancer Res. 1977;37:1719–1726. PubMed
Mattarollo SR, Loi S, Duret H, Ma Y, Zitvogel L, Smyth MJ. Pivotal role of innate and adaptive immunity in anthracycline chemotherapy of established tumors. Cancer Res American Association for Cancer Research. 2011;71:4809–4820. PubMed
Bloy N, Garcia P, Laumont CM, Pitt JM, Sistigu A, Stoll G, et al. Immunogenic stress and death of cancer cells: contribution of antigenicity vs adjuvanticity to immunosurveillance. Immunol Rev. 2017;280:165–174. doi: 10.1111/imr.12582. PubMed DOI
Galluzzi L, Bravo-San Pedro JM, Demaria S, Formenti SC, Kroemer G. Activating autophagy to potentiate immunogenic chemotherapy and radiation therapy. Nat Rev Clin Oncol. 2017;14:247–258. doi: 10.1038/nrclinonc.2016.183. PubMed DOI
Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu Rev Immunol Annual Reviews. 2013;31:51–72. doi: 10.1146/annurev-immunol-032712-100008. PubMed DOI
Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P, Vandenabeele P. Immunogenic cell death and DAMPs in cancer therapy. Nat Rev Cancer Nature Publishing Group. 2012;12:860–875. doi: 10.1038/nrc3380. PubMed DOI
Martin K, Müller P, Schreiner J, Prince SS, Lardinois D, Heinzelmann-Schwarz VA, et al. The microtubule-depolymerizing agent ansamitocin P3 programs dendritic cells toward enhanced anti-tumor immunity. Cancer Immunol Immunother. Springer Berlin Heidelberg. 2014;63:925–938. doi: 10.1007/s00262-014-1565-4. PubMed DOI PMC
Müller P, Martin K, Theurich S, Schreiner J, Savic S, Terszowski G, et al. Microtubule-depolymerizing agents used in antibody-drug conjugates induce antitumor immunity by stimulation of dendritic cells. Cancer Immunol Res. 2014;2:741–755. doi: 10.1158/2326-6066.CIR-13-0198. PubMed DOI
Waldmeier L, Hellmann I, Gutknecht CK, Wolter FI, Cook SC, Reddy ST, et al. Transpo-mAb display: Transposition-mediated B cell display and functional screening of full-length IgG antibody libraries. mAbs. Taylor & Francis. 2016;8:726–740. PubMed PMC
Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. BioMed Central. 2014;15:550. doi: 10.1186/s13059-014-0550-8. PubMed DOI PMC
Huber W, Carey VJ, Gentleman R, Anders S, Carlson M, Carvalho BS, et al. Orchestrating high-throughput genomic analysis with Bioconductor. Nature Methods Nature Publishing Group. 2015;12:115–121. doi: 10.1038/nmeth.3252. PubMed DOI PMC
Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res 2015;43:e47–7. PubMed PMC
Bolotin DA, Poslavsky S, Davydov AN, Frenkel FE, Fanchi L, Zolotareva OI, et al. Antigen receptor repertoire profiling from RNA-seq data. Nat Biotechnol. 2017;35:908–911. doi: 10.1038/nbt.3979. PubMed DOI PMC
Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013;39:1–10. doi: 10.1016/j.immuni.2013.07.012. PubMed DOI
Galon J, Mlecnik B, Bindea G, Angell HK, Berger A, Lagorce C, et al. Towards the introduction of the “Immunoscore” in the classification of malignant tumours. Jubb A, Koeppen H, Reis-Filho J, editors. J Pathol. John Wiley & Sons, Ltd; 2014;232:199–209. PubMed PMC
de Melo GD, Jardim DLF, Marchesi MSP, Hortobagyi GN. Mechanisms of resistance and sensitivity to anti-HER2 therapies in HER2+ breast cancer. Oncotarget Impact Journals. 2016;7:64431–64446. PubMed PMC
Ríos-Luci C, García-Alonso S, Díaz-Rodríguez E, Nadal-Serrano M, Arribas J, Ocaña A, et al. Resistance to the Antibody-drug conjugate T-DM1 is based in a reduction in lysosomal proteolytic activity. Cancer Res. 2017;77:4639–4651. doi: 10.1158/0008-5472.CAN-16-3127. PubMed DOI
Rios-Doria J, Harper J, Rothstein R, Wetzel L, Chesebrough J, Marrero A, et al. Antibody-drug conjugates bearing Pyrrolobenzodiazepine or Tubulysin payloads are immunomodulatory and synergize with multiple immunotherapies. Cancer Res. American Association for Cancer Research. 2017;77:2686–2698. PubMed
Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359:1350–1355. doi: 10.1126/science.aar4060. PubMed DOI PMC
Pfirschke C, Engblom C, Rickelt S, Cortez-Retamozo V, Garris C, Pucci F, et al. Immunogenic chemotherapy sensitizes tumors to checkpoint blockade therapy. Immunity. 2016;44:343–354. doi: 10.1016/j.immuni.2015.11.024. PubMed DOI PMC
Detection of immunogenic cell death and its relevance for cancer therapy