Dynamics of cytotoxic T cell subsets during immunotherapy predicts outcome in acute myeloid leukemia

Frida Ewald Sander, Anna Rydström, Elin Bernson, Roberta Kiffin, Rebecca Riise, Johan Aurelius, Harald Anderson, Mats Brune, Robin Foà, Kristoffer Hellstrand, Fredrik B Thorén, Anna Martner, Frida Ewald Sander, Anna Rydström, Elin Bernson, Roberta Kiffin, Rebecca Riise, Johan Aurelius, Harald Anderson, Mats Brune, Robin Foà, Kristoffer Hellstrand, Fredrik B Thorén, Anna Martner

Abstract

Preventing relapse after chemotherapy remains a challenge in acute myeloid leukemia (AML). Eighty-four non-transplanted AML patients in first complete remission received relapse-preventive immunotherapy with histamine dihydrochloride and low-dose interleukin-2 in an international phase IV trial (ClinicalTrials.gov; NCT01347996). Blood samples were drawn during cycles of immunotherapy and analyzed for CD8+ (cytotoxic) T cell phenotypes in blood. During the first cycle of therapy, a re-distribution of cytotoxic T cells was observed comprising a reduction of T effector memory cells and a concomitant increase of T effector cells. The dynamics of T cell subtypes during immunotherapy prognosticated relapse and survival, in particular among older patients and remained significantly predictive of clinical outcome after correction for potential confounders. Presence of CD8+ T cells with specificity for leukemia-associated antigens identified patients with low relapse risk. Our results point to novel aspects of T cell-mediated immunosurveillance in AML and provide conceivable biomarkers in relapse-preventive immunotherapy.

Keywords: Immune response; Immunity; Immunology and Microbiology Section; acute myeloid leukemia; antigen-specific T cells; cytotoxic T cells; immunotherapy.

Conflict of interest statement

CONFLICTS OF INTEREST

Authors MB and KH are past or present consultants to the study sponsor (Meda Pharma). Author KH holds patents protecting the use of histamine dihydrochloride in cancer immunotherapy. Authors AM, RF and FBT have received honoraria and/or travel grants from the study sponsor. The other authors declare no conflict of interest.

Figures

Figure 1. Overview of the Re:Mission phase…
Figure 1. Overview of the Re:Mission phase IV trial
Eligible AML patients in first complete remission (CR) received ten 3-week cycles of HDC/IL-2 over 18 months. Peripheral blood mononuclear cells (PBMC) were isolated from blood collected before and after cycles 1 and 3. Patients were followed-up for 6 months after completing the last treatment cycle.
Figure 2. Distribution of CD8 + subsets…
Figure 2. Distribution of CD8+ subsets in non-relapsing and relapsing AML patients during immunotherapy with HDC/IL-2
A. Blood counts of CD8+ T cells before (D1) and after (D21) the first and third cycles of HDC/IL-2 treatment (C1D1 n = 62; C1D21 n = 54; C3D1 n = 52; C3D21 n = 51). B. Gating strategy for determining naïve (TN; CD45RA+CCR7+), central memory (TCM; CD45RO+CCR7+), effector memory (TEM; CD45RO+CCR7−) and effector (Teff; CD45RA+CCR7−) cells within the CD8+ T cell compartment. C-F. Frequency of the CD8+ subpopulations TNC., TCMD., TEME. and TeffF. cells in non-relapsing (n = 18) and relapsing (n = 26) patients at the onset (C1D1) or end of (C1D21) the first cycle of immunotherapy. Statistical analysis was performed by Student's paired t-test.
Figure 3. Impact of altered distribution of…
Figure 3. Impact of altered distribution of CD8+subsets on the clinical outcome of patients receiving HDC/IL-2
In A-D. all patients, and in F-I. patients ≥ 60 years old, were dichotomized based on induction or reduction of the frequency of CD8+ T cell subsets during the first treatment cycle, followed by analyses of LFS and OS by the logrank test. In E. all patients, and in J. patients≥ 60 years old, were dichotomized based on transition (trans) or no transition from TEM to Teff cells and LFS and OS were analyzed by the logrank test. A patient was considered transition-positive by the occurrence of a reduction of TEM cells (%) and a simultaneous induction of Teff cells (%) during the first treatment cycle.
Figure 4. Impact of HLA-DR expression and…
Figure 4. Impact of HLA-DR expression and leukemia-specific CD8+T cells on LFS in patients receiving HDC/IL-2
A. Patients were dichotomized by the median HLA-DR expression on CD3+CD8+ T cells at onset of therapy (C1D1; n = 44) or after the first treatment cycle (C1D21; n = 47). LFS and OS were analyzed by the logrank test. B-C. Blood samples from patients undergoing HDC/IL-2 treatment were stimulated with a pool of peptides from leukemia-associated antigens (AML-peptides) or a pool of peptides from CMV, EBV and influenza viruses (CEF-peptides), or no peptides (negative control). The percentage of IFN-γ producing CD8+ T cells was determined by flow cytometry. In B. representative dot plots show IFN-γ production in samples without stimulation and samples stimulated with AML- or CEF-peptides. In C. patients were dichotomized based on the presence or absence of AML-specific or CEF-specific CD8+ T cells, followed by analysis of LFS by the logrank test. Only patients with no events occurring before the last time point of analysis of antigen-specific T cells (C3D21; 105 days) were considered in the latter analyses.
Figure 5. Impact of T EM to…
Figure 5. Impact of TEM to Teff cell transition and NK cell NKp46 expression on clinical outcome
A.-B. Patients were regarded as transition-positive when showing a reduction of TEM cells (%) and a simultaneous induction of Teff cells (%) during the first treatment cycle, and were considered NKp46high when their CD16+ NK cells expressed above median levels of NKp46 after the first cycle of immunotherapy (C1D21). Data show the LFS and OS (analyzed by the logrank test for trend) of patients with transition TEM-Teff and NKp46high (both), transition only, NKp46high only, no transition TEM-Teff and low NKp46 expression (neither).

References

    1. Burnett AK, Goldstone A, Hills RK, Milligan D, Prentice A, Yin J, Wheatley K, Hunter A, Russell N. Curability of patients with acute myeloid leukemia who did not undergo transplantation in first remission. Journal of clinical oncology. 2013;31:1293–1301.
    1. Martner A, Thoren FB, Aurelius J, Hellstrand K. Immunotherapeutic strategies for relapse control in acute myeloid leukemia. Blood reviews. 2013;27:209–216.
    1. Appelbaum FR. Haematopoietic cell transplantation as immunotherapy. Nature. 2001;411:385–389.
    1. Kolb HJ. Graft-versus-leukemia effects of transplantation and donor lymphocytes. Blood. 2008;112:4371–4383.
    1. Montagna D, Maccario R, Locatelli F, Montini E, Pagani S, Bonetti F, Daudt L, Turin I, Lisini D, Garavaglia C, Dellabona P, Casorati G. Emergence of antitumor cytolytic T cells is associated with maintenance of hematologic remission in children with acute myeloid leukemia. Blood. 2006;108:3843–3850.
    1. Greiner J, Schmitt M, Li L, Giannopoulos K, Bosch K, Schmitt A, Dohner K, Schlenk RF, Pollack JR, Dohner H, Bullinger L. Expression of tumor-associated antigens in acute myeloid leukemia: Implications for specific immunotherapeutic approaches. Blood. 2006;108:4109–4117.
    1. Pizzitola I, Anjos-Afonso F, Rouault-Pierre K, Lassailly F, Tettamanti S, Spinelli O, Biondi A, Biagi E, Bonnet D. Chimeric antigen receptors against CD33/CD123 antigens efficiently target primary acute myeloid leukemia cells in vivo. Leukemia. 2014;28:1596–1605.
    1. Van Tendeloo VF, Van de Velde A, Van Driessche A, Cools N, Anguille S, Ladell K, Gostick E, Vermeulen K, Pieters K, Nijs G, Stein B, Smits EL, Schroyens WA, Gadisseur AP, Vrelust I, Jorens PG, et al. Induction of complete and molecular remissions in acute myeloid leukemia by Wilms' tumor 1 antigen-targeted dendritic cell vaccination. Proceedings of the National Academy of Sciences of the United States of America. 2010;107:13824–13829.
    1. Krupka C, Kufer P, Kischel R, Zugmaier G, Lichtenegger FS, Kohnke T, Vick B, Jeremias I, Metzeler KH, Altmann T, Schneider S, Fiegl M, Spiekermann K, Bauerle PA, Hiddemann W, Riethmuller G, et al. Blockade of the PD-1/PD-L1 axis augments lysis of AML cells by the CD33/CD3 BiTE antibody construct AMG 330: reversing a T-cell-induced immune escape mechanism. Leukemia. 2016;30:484–91.
    1. Brune M, Castaigne S, Catalano J, Gehlsen K, Ho AD, Hofmann WK, Hogge DE, Nilsson B, Or R, Romero AI, Rowe JM, Simonsson B, Spearing R, Stadtmauer EA, Szer J, Wallhult E, et al. Improved leukemia-free survival after postconsolidation immunotherapy with histamine dihydrochloride and interleukin-2 in acute myeloid leukemia: results of a randomized phase 3 trial. Blood. 2006;108:88–96.
    1. Farber DL, Yudanin NA, Restifo NP. Human memory T cells: generation, compartmentalization and homeostasis. Nat Rev Immunol. 2014;14:24–35.
    1. Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu Rev Immunol. 2004;22:745–763.
    1. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015;348:62–68.
    1. Restifo NP, Gattinoni L. Lineage relationship of effector and memory T cells. Current opinion in immunology. 2013;25:556–563.
    1. Roychoudhuri R, Lefebvre F, Honda M, Pan L, Ji Y, Klebanoff CA, Nichols CN, Fourati S, Hegazy AN, Goulet JP, Gattinoni L, Nabel GJ, Gilliet M, Cameron M, Restifo NP, Sekaly RP, et al. Transcriptional profiles reveal a stepwise developmental program of memory CD8(+) T cell differentiation. Vaccine. 2015;33:914–923.
    1. Burnett AK. Treatment of acute myeloid leukemia: are we making progress? ASH Education Program Book. 2012;2012:1–6.
    1. Liao W, Lin JX, Leonard WJ. IL-2 family cytokines: new insights into the complex roles of IL-2 as a broad regulator of T helper cell differentiation. Current opinion in immunology. 2011;23:598–604.
    1. Scheibenbogen C, Letsch A, Thiel E, Schmittel A, Mailaender V, Baerwolf S, Nagorsen D, Keilholz U. CD8 T-cell responses to Wilms tumor gene product WT1 and proteinase 3 in patients with acute myeloid leukemia. Blood. 2002;100:2132–2137.
    1. Greiner J, Schneider V, Schmitt M, Gotz M, Dohner K, Wiesneth M, Dohner H, Hofmann S. Immune responses against the mutated region of cytoplasmatic NPM1 might contribute to the favorable clinical outcome of AML patients with NPM1 mutations (NPM1mut) Blood. 2013;122:1087–1088.
    1. Ko HS, Fu SM, Winchester RJ, Yu DT, Kunkel HG. Ia determinants on stimulated human T lymphocytes. Occurrence on mitogen- and antigen-activated T cells. J Exp Med. 1979;150:246–255.
    1. Arruvito L, Payaslian F, Baz P, Podhorzer A, Billordo A, Pandolfi J, Semeniuk G, Arribalzaga E, Fainboim L. Identification and clinical relevance of naturally occurring human CD8+HLA-DR+ regulatory T cells. J Immunol. 2014;193:4469–4476.
    1. Imamichi H, Lempicki RA, Adelsberger JW, Hasley RB, Rosenberg A, Roby G, Rehm CA, Nelson A, Krishnan S, Pavlick M, Woods CJ, Baseler MW, Lane HC. The CD8+ HLA-DR+ T cells expanded in HIV-1 infection are qualitatively identical to those from healthy controls. Eur J Immunol. 2012;42:2608–2620.
    1. Thoren FB, Romero AI, Brune M, Hellstrand K. Histamine dihydrochloride and low-dose interleukin-2 as post-consolidation immunotherapy in acute myeloid leukemia. Expert Opin Biol Ther. 2009;9:1217–1223.
    1. Martner A, Rydstrom A, Riise RE, Aurelius J, Anderson H, Brune M, Foa R, Hellstrand K, Thoren FB. Role of natural killer cell subsets and natural cytotoxicity receptors for the outcome of immunotherapy in acute myeloid leukemia. Oncoimmunology. 2015 doi: 10.1080/2162402X.2162015.1041701.
    1. Martner A, Rydstrom A, Riise RE, Aurelius J, Anderson H, Brune M, Foa R, Hellstrand K, Thoren FB. NK cell expression of natural cytotoxicity receptors may determine relapse risk in older AML patients undergoing immunotherapy for remission maintenance. Oncotarget. 2015 doi: 10.18632/oncotarget.15559.
    1. Dohner H, Estey EH, Amadori S, Appelbaum FR, Buchner T, Burnett AK, Dombret H, Fenaux P, Grimwade D, Larson RA, Lo-Coco F, Naoe T, Niederwieser D, Ossenkoppele GJ, Sanz MA, Sierra J, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115:453–474.

Source: PubMed

3
Sottoscrivi