We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Article

Eosinophilic count as a biomarker for prognosis of melanoma patients and its importance in the response to immunotherapy

    Alvaro Moreira

    Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany

    ,
    Waltraud Leisgang

    Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany

    ,
    Gerold Schuler

    Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany

    &
    Lucie Heinzerling

    *Author for correspondence:

    E-mail Address: lucie.heinzerling@uk-erlangen.de

    Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany

    Published Online:https://doi.org/10.2217/imt-2016-0138

    Aim: The prognostic role of eosinophils in cancer has been controversial. Some entities such as gastrointestinal cancers show a better survival, while others such as Hodgkin's lymphoma a worse survival in patients with eosinophilia. Patients who exhibited an increase in eosinophils upon therapy with ipilimumab or pembrolizumab were shown to survive longer. We wanted to investigate whether eosinophilia is a prognostic marker in metastatic melanoma. Methods: In total, 173 patients with metastatic melanoma from our data base (median age 60 years; n = 86 with immunotherapy, n = 87 without immunotherapy) were analyzed for eosinophil counts and survival over the course of 12 years. Eosinophilic count was detected by peripheral blood smear. The ethical committee had approved this retrospective study. Results: Melanoma patients with eosinophilia at any point in their course of disease show a trend toward longer survival independently of their therapy. There is a statistically significant difference for the patients who survive at least 12 months (p < 0.005). In patients with checkpoint inhibitor therapy, survival was significantly prolonged in every patient with eosinophilia (p < 0.05). Furthermore, 69% of the patients treated with immunotherapy experienced at least once an eosinophilia of 5% or greater compared with 46% in the immunotherapy naive-group; for an eosinophilia of 10% values were 30 and 9%, respectively. Interestingly, in patients with more than 20% eosinophils (n = 7) survival was prolonged with a median of 35 months (range 19–60 months) as compared with 16 months (range 1–117 months). Conclusion: Eosinophilia is a prognostic marker in patients with metastatic melanoma.

    References

    • 1 Simson L, Ellyard JI, Dent LA et al. Regulation of carcinogenesis by IL-5 and CCL11: a potential role for eosinophils in tumor immune surveillance. J. Immunol. 178(7), 4222–4229 (2007).
    • 2 Costain DJ, Guha AK, Liwski RS, Lee TD. Murine hypodense eosinophils induce tumour cell apoptosis by a granzyme B-dependent mechanism. Cancer Immunol. Immunother. 50(6), 293–299 (2001).
    • 3 Ikutani M, Yanagibashi T, Ogasawara M et al. Identification of innate IL-5-producing cells and their role in lung eosinophil regulation and antitumor immunity. J. Immunol. 188(2), 703–713 (2012).
    • 4 Legrand F, Driss V, Delbeke M et al. Human eosinophils exert TNF-alpha and granzyme A-mediated tumoricidal activity toward colon carcinoma cells. J. Immunol. 185(12), 7443–7451 (2010).
    • 5 Lowe D, Jorizzo J, Hutt MS. Tumour-associated eosinophilia: a review. J. Clin. Pathol. 34(12), 1343–1348 (1981).
    • 6 Samoszuk M. Eosinophils and human cancer. Histol. Histopathol. 12(3), 807–812 (1997).
    • 7 Ohashi Y, Ishibashi S, Suzuki T et al. Significance of tumor associated tissue eosinophilia and other inflammatory cell infiltrate in early esophageal squamous cell carcinoma. Anticancer Res. 20(5A), 3025–3030 (2000).
    • 8 Samoszuk M, Deng T, Hamamura MJ, Su MY, Asbrock N, Nalcioglu O. Increased blood clotting, microvascular density, and inflammation in eotaxin-secreting tumors implanted into mice. Am. J. Pathol. 165(2), 449–456 (2004).
    • 9 Lotfi R, Lee JJ, Lotze MT. Eosinophilic granulocytes and damage-associated molecular pattern molecules (DAMPs): role in the inflammatory response within tumors. J. Immunother. 30(1), 16–28 (2007).
    • 10 Davis BP, Rothenberg ME. Eosinophils and cancer. Cancer Immunol. Res. 2(1), 1–8 (2014).
    • 11 Gatault S, Legrand F, Delbeke M, Loiseau S, Capron M. Involvement of eosinophils in the anti-tumor response. Cancer Immunol. Immunother. 61(9), 1527–1534 (2012).
    • 12 Prizment AE, Vierkant RA, Smyrk TC et al. Tumor eosinophil infiltration and improved survival of colorectal cancer patients: Iowa Women's Health Study. Mod. Pathol. 29(5), 516–527 (2016).
    • 13 Sakkal S, Miller S, Apostolopoulos V, Nurgali K. Eosinophils in cancer: favourable or unfavourable? Curr. Med. Chem. 23(7), 650–666 (2016).
    • 14 Minton K. Granulocytes: eosinophils enable the antitumour T cell response. Nat. Rev. Immunol. 15(6), 333 (2015).
    • 15 Carretero R, Sektioglu IM, Garbi N, Salgado OC, Beckhove P, Hammerling GJ. Eosinophils orchestrate cancer rejection by normalizing tumor vessels and enhancing infiltration of CD8(+) T cells. Nat. Immunol. 16(6), 609–617 (2015).
    • 16 Cormier SA, Taranova AG, Bedient C et al. Pivotal Advance: eosinophil infiltration of solid tumors is an early and persistent inflammatory host response. J. Leukoc. Biol. 79(6), 1131–1139 (2006).
    • 17 Capobianco A, Manfredi AA, Monno A, Rovere-Querini P, Rugarli C. Melanoma and lymphoma rejection associated with eosinophil infiltration upon intratumoral injection of dendritic and NK/LAK cells. J. Immunother. 31(5), 458–465 (2008).
    • 18 Lipson EJ, Sharfman WH, Chen S et al. Safety and immunologic correlates of Melanoma GVAX, a GM-CSF secreting allogeneic melanoma cell vaccine administered in the adjuvant setting. J. Transl. Med. 13, 214 (2015).
    • 19 Delyon J, Mateus C, Lefeuvre D et al. Experience in daily practice with ipilimumab for the treatment of patients with metastatic melanoma: an early increase in lymphocyte and eosinophil counts is associated with improved survival. Ann. Oncol. 24(6), 1697–1703 (2013).
    • 20 Umansky V, Utikal J, Gebhardt C. Predictive immune markers in advanced melanoma patients treated with ipilimumab. Oncoimmunology 5(6), e1158901 (2016).
    • 21 Weide B, Martens A, Hassel JC et al. Baseline biomarkers for outcome of melanoma patients treated with pembrolizumab. Clin. Cancer Res. doi:10.1158/1078-0432.ccr-16-0127 (2016) (Epub ahead of print).
    • 22 Groß S, Erdmann M, Schuler G. Dendritic cell vaccination on the long run: 11-year survival in advanced melanoma patients is equivalent to ipilimumab-treatment (2016) (Submitted).
    • 23 Rodgers S, Rees RC, Hancock BW. Changes in the phenotypic characteristics of eosinophils from patients receiving recombinant human interleukin-2 (rhIL-2) therapy. Br. J. Haematol. 86(4), 746–753 (1994).
    • 24 Schaefer JT, Patterson JW, Deacon DH et al. Dynamic changes in cellular infiltrates with repeated cutaneous vaccination: a histologic and immunophenotypic analysis. J. Transl. Med. 8, 79 (2010).
    • 25 Faries MB, Hsueh EC, Ye X, Hoban M, Morton DL. Effect of granulocyte/macrophage colony-stimulating factor on vaccination with an allogeneic whole-cell melanoma vaccine. Clin. Cancer Res. 15(22), 7029–7035 (2009).
    • 26 Mackensen A, Meidenbauer N, Vogl S, Laumer M, Berger J, Andreesen R. Phase I study of adoptive T-cell therapy using antigen-specific CD8+ T cells for the treatment of patients with metastatic melanoma. J. Clin. Oncol. 24(31), 5060–5069 (2006).
    • 27 Sedgwick JB, Frick WE, Sondel PM, Hank JA, Borden E, Busse WW. The appearance of hypodense eosinophils during interleukin-2 treatment. J. Allergy Clin. Immunol. 85(3), 557–566 (1990).
    • 28 Rivoltini L, Viggiano V, Spinazze S et al. In vitro anti-tumor activity of eosinophils from cancer patients treated with subcutaneous administration of interleukin 2. Role of interleukin 5. Int. J. Cancer 54(1), 8–15 (1993).
    • 29 Phan GQ, Yang JC, Sherry RM et al. Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proc. Natl Acad. Sci. USA 100(14), 8372–8377 (2003).
    • 30 Attia P, Phan GQ, Maker AV et al. Autoimmunity correlates with tumor regression in patients with metastatic melanoma treated with anti-cytotoxic T-lymphocyte antigen-4. J. Clin. Oncol. 23(25), 6043–6053 (2005).
    • 31 Brahmer JR, Drake CG, Wollner I et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J. Clin. Oncol. 28(19), 3167–3175 (2010).
    • 32 Hamid O, Robert C, Daud A et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N. Engl. J. Med. 369(2), 134–144 (2013).
    • 33 Gebhardt C, Sevko A, Jiang H et al. Myeloid cells and related chronic inflammatory factors as novel predictive markers in melanoma treatment with ipilimumab. Clin. Cancer Res. 21(24), 5453–5459 (2015).