Pre-existing effector T-cell levels and augmented myeloid cell composition denote response to CDK4/6 inhibitor palbociclib and pembrolizumab in hormone receptor-positive metastatic breast cancer

Colt Egelston, Weihua Guo, Susan Yost, Jin Sun Lee, David Rose, Christian Avalos, Jian Ye, Paul Frankel, Daniel Schmolze, James Waisman, Peter Lee, Yuan Yuan, Colt Egelston, Weihua Guo, Susan Yost, Jin Sun Lee, David Rose, Christian Avalos, Jian Ye, Paul Frankel, Daniel Schmolze, James Waisman, Peter Lee, Yuan Yuan

Abstract

Background: Single-agent pembrolizumab treatment of hormone receptor-positive metastatic breast cancer (MBC) has demonstrated modest clinical responses. Little is known about potential biomarkers or mechanisms of response to immune checkpoint inhibitors (ICIs) in patients with HR+ MBC. The present study presents novel immune correlates of clinical responses to combined treatment with CDK4/6i and ICI.

Methods: A combined analysis of two independent phase I clinical trials treating patients with HR+ MBC was performed. Patients treated with the combination of the CDK4/6i palbociclib+the ICI pembrolizumab+the aromatase inhibitor (AI) letrozole (palbo+pembro+AI) were compared with patients treated with pembrolizumab+AI (pembro+AI). Peripheral blood mononuclear cells collected at pretreatment, 3 weeks (cycle 2 day 1) and 9 weeks (cycle 4 day 1) were characterized by high-parameter flow cytometry to assess baseline immune subset composition and longitudinal changes in response to therapy.

Results: In the peripheral blood, higher pretreatment frequencies of effector memory CD45RA+ CD8+ T cells and effector memory CD4+ T cells were observed in responders to palbo+pembro+AI. In contrast, this was not observed in pembro+AI-treated patients. We further characterized T-cell subsets of effector-like killer cell lectin-like receptor subfamily G member 1 (KLRG1+) ICOS+ CD4+ T cells and KLRG1+ CD45RA+ CD8+ T cells as baseline biomarkers of response. In comparison, pretreatment levels of tumor-infiltrating lymphocyte, tumor mutation burden, tumor programmed death-ligand 1 expression, and overall immune composition did not associate with clinical responses. Over the course of treatment, significant shifts in myeloid cell composition and phenotype were observed in palbo+pembro+AI-treated patients, but not in those treated with pembro+AI. We identified increased fractions of type 1 conventional dendritic cells (cDC1s) within circulating dendritic cells and decreased classical monocytes (cMO) within circulating monocytes only in patients treated with palbociclib. We also demonstrated that in palbociclib-treated patients, cDC1 and cMO displayed increased CD83 and human leukocyte antigen-DR isotype (HLA-DR) expression, respectively, suggesting increased maturation and antigen presentation capacity.

Conclusions: Pre-existing circulating effector CD8+ and CD4+ T cells and dynamic modulation of circulating myeloid cell composition denote response to combined pembrolizumab and palbociclib therapy for patients with HR+ MBC.

Trial registration number: NCT02778685 and NCI02648477.

Keywords: CD4-positive T-lymphocytes; CD8-positive T-lymphocytes; breast neoplasms; immunotherapy.

Conflict of interest statement

Competing interests: YY has contracted clinical trials and research projects sponsored by Merck, Eisai, Novartis, Puma, Genentech, and Pfizer. She is a consultant for Puma and is on the Speakers Bureau for Eisai. There are no patents or products in development or marketed products associated with this research to declare. There are no restrictions on sharing of data and/or materials.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Baseline tumor and peripheral blood characterization of HR+ MBC responders and non-responders to palbo+pembro+AI. Baseline tumor biopsies were assessed for TILs (A), PD-L1 expression (B, reported as fraction of each cohort with PD-L1+ scored tumors), and tumor mutation burden (C, reported as m/MB). Baseline peripheral blood samples were examined by flow cytometry for lymphocyte composition frequencies, including CD4+ T cells, CD8+ T cells, CD3+ CD56+ NKTs, CD19+ B cells, or CD3− CD56+ NKs (D). The composition of NKs was further assessed for CD56bright, CD16bright, and CD56dim CD16dim NK subsets (E). Monocyte subsets were assessed as a fraction of a myeloid cell gate for frequencies of CD14+ CD16− cMOs, CD14+CD16+ iMOs, and CD14− CD16+ ncMOs (F). Dendritic cell subsets were similarly assessed as a fraction of a myeloid cell gate for frequencies of CD141+ dendritic cells (cDC1), CD1c+ dendritic cells (cDC2), and CD123+ pDCs (G). NR patients are depicted in blue; responder (R) patients are depicted in red. (D) NR, n=7; R, n=9. (E–G) NR, n=7; R, n=10. AI, aromatase inhibitor; cDC, conventional dendritic cell; cDC1, type 1 conventional dendritic cell; cDC2, type 2 conventional dendritic cell; cMO, classical monocyte; HR, hormone receptor; iMO, intermediate monocyte; m/MB, mutations/megabase; MBC, metastatic breast cancer; ncMO, non-classical monocyte; NK, natural killer; NKT, natural killer T cell; NR, non-responder; palbo, palbociclib; pDC, plasmacytoid dendritic cell; PD-L1, programmed death-ligand 1; pembro, pembrolizumab; R, responder; TIL, tumor-infiltrating lymphocyte; TMB, tumor mutation burden.
Figure 2
Figure 2
Distinct pre-existing effector T-cell subsets in responders to palbo+pembro+ AI. CD8+ T cells (A) and CD4+ T cells (B) were further assessed by flow cytometry for naïve (CCR7+ and CD45RA+), CM (CCR7+ and CD45RA−), EM (CCR7− and CD45RA−), and EmrA (CCR7− and CD45RA+) cell subsets. Dimensionality reduction by the FlowSOM algorithm was performed to identify T-cell metaclusters in an unbiased manner as shown in t-distributed stochastic neighbor embedding (tSNE) projections of R and NR patients (C). A heatmap of identified clusters displays expression of various surface proteins used for each identified cell cluster. Percentages of identified non-naïve CD8+ T-cell clusters as a fraction of total CD8+ T cells are shown in NR and R patients (D). Percentages of identified non-naïve CD4+ T-cell clusters as a fraction of total CD4+ T cells are shown in NR and R patients (E). Percentages of non-classical CD4+ CD8+ DP cells and CD8dim cells as a fraction of total lymphocytes (F). *P<0.05, **P<0.01; unpaired, two-tailed t-tests. NR patients are depicted in blue (n=7) and R patients are depicted in red (n=9). AI, aromatase inhibitor; CM, central memory; DP, double-positive; EM, effector memory; EMRA, effector memory CD45RA+; NR, non-responder; palbo, palbociclib; pembro, pembrolizumab; R, responder.
Figure 3
Figure 3
Peripheral immune composition changes in Rs to palbo+pembro+ AI compared with NRs to pembro+AI. Baseline immune subset composition in responders to palbo+pembro+AI treatment (red) was compared with non-responders to pembro+AI treatment (green). Major lymphocyte subsets (A), CD8+ T-cell memory subsets (B), CD4+ T-cell memory subsets (C), NK subsets (D), monocyte subsets (E), and dendritic cell subsets (F) were assessed by flow cytometry. Changes in immune composition from baseline to C2D1 were also assessed by flow cytometry for all major non-T-cell immune subsets (G) and T-cell clusters previously identified (H) by calculating fold change from baseline. Log2 transformed fold changes are depicted and populations are sorted from least significant difference (top) to most significant difference (bottom). C2D1 palbo+pembro+AI (gold); C2D1 pembro+AI (green). *P<0.05, **P<0.01; unpaired, two-tailed t-tests. (A–C) R: palbo+pembro+AI, n=9; NR: pembro+AI, n=10. (DcF) R: palbo+pembro+AI, n=10; NR: pembro AI, n=10. (G–H) palbo+pembro+AI, n=14; pembro+AI, n=10. AI, aromatase inhibitor; C2D1, cycle 2 day 1; cDC1, type 1 conventional dendritic cell; cDC2, type 2 conventional dendritic cell; CM, central memory; cMO, classical monocyte; EM, effector memory; EMRA, effector memory CD45RA+; iMO, intermediate monocyte; ncMO, non-classical monocyte; NK, natural killer cell; NKT, natural killer T cell; NR, non-responder; palbo, palbociclib; pDC, plasmacytoid dendritic cell; pembro, pembrolizumab; R, responder.
Figure 4
Figure 4
Dynamic peripheral myeloid cell changes in patients treated with palbo+pembro+AI compared with pembro+AI Changes in cell population percentages over the course of therapy were compared between palbo+pembro+AI-treated and pembro+AI-treated patients. Changes in frequencies of cDC1 (A), cDC2 (B), pDC (C), cMO (D), iMO (E), and ncMO (F) were evaluated as fractions of myeloid cell gates. (A–F) Data are normalized by subtraction of cell population percentages identified at baseline in the same patient. cDC1 frequencies among total circulating dendritic cells (cDC1+cDC2) are shown over the course of treatment (G). cMO frequencies among total circulating monocytes (cMO+iMO+ncMO) are shown over the course of treatment (H). Baseline palbo+pembro+AI depicted by gray open circle (n=17); C2D1 palbo pembro+AI depicted in gold (n=14); C4D1 palbo+pembro+AI depicted in purple (n=13), C2D1 pembro+AI depicted in green (n=10). *P

Figure 5

Palbociclib yields increased frequencies of…

Figure 5

Palbociclib yields increased frequencies of circulating mature cDC1 and cMOs. Circulating dendritic cell…

Figure 5
Palbociclib yields increased frequencies of circulating mature cDC1 and cMOs. Circulating dendritic cell and monocyte subsets were assessed by flow cytometry for changes in maturation status over the course of therapy. Changes in CD83 expression compared with baseline pretreatment were examined in cDC1 (A) and cDC2 (B) cells. Monocyte subsets were examined for changes in frequencies of human leukocyte antigen-DR isotype (HLA-DR)high cells compared with baseline (C). Changes in HLA-DR expression as measured by change in MFI relative to baseline MFI on the same cell subsets was assessed in cMO (D), iMO (E), and ncMO (F). Representative histograms are shown, with the dotted line displaying matched baseline cell subset marker expression. Baseline palbo+pembro+AI depicted by gray open circle (n=17); C2D1 palbo+pembro+AI depicted in gold (n=14); C4D1 palbo+pembro+AI depicted in purple (n=13), C2D1pembro+AI depicted in green (n=10). *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001; Dunnett’s multiple comparisons test. AI, aromatase inhibitor; C2D1; cycle 2 day 1; C4D1, cycle 4 day 1; cDC1, type 1 conventional dendritic cell; cDC2, type 2 conventional dendritic cell; cMO, classical monocyte; iMO, intermediate monocyte; MFI, median fluorescent intensity; ncMO, non-classical monocyte; palbo, palbociclib; pDC, plasmacytoid dendritic cell; pembro, pembrolizumab.
Figure 5
Figure 5
Palbociclib yields increased frequencies of circulating mature cDC1 and cMOs. Circulating dendritic cell and monocyte subsets were assessed by flow cytometry for changes in maturation status over the course of therapy. Changes in CD83 expression compared with baseline pretreatment were examined in cDC1 (A) and cDC2 (B) cells. Monocyte subsets were examined for changes in frequencies of human leukocyte antigen-DR isotype (HLA-DR)high cells compared with baseline (C). Changes in HLA-DR expression as measured by change in MFI relative to baseline MFI on the same cell subsets was assessed in cMO (D), iMO (E), and ncMO (F). Representative histograms are shown, with the dotted line displaying matched baseline cell subset marker expression. Baseline palbo+pembro+AI depicted by gray open circle (n=17); C2D1 palbo+pembro+AI depicted in gold (n=14); C4D1 palbo+pembro+AI depicted in purple (n=13), C2D1pembro+AI depicted in green (n=10). *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001; Dunnett’s multiple comparisons test. AI, aromatase inhibitor; C2D1; cycle 2 day 1; C4D1, cycle 4 day 1; cDC1, type 1 conventional dendritic cell; cDC2, type 2 conventional dendritic cell; cMO, classical monocyte; iMO, intermediate monocyte; MFI, median fluorescent intensity; ncMO, non-classical monocyte; palbo, palbociclib; pDC, plasmacytoid dendritic cell; pembro, pembrolizumab.

References

    1. Narayan P, Wahby S, Gao JJ, et al. . Fda approval summary: Atezolizumab plus paclitaxel protein-bound for the treatment of patients with advanced or metastatic TNBC whose tumors express PD-L1. Clin Cancer Res 2020;26:2284–9. 10.1158/1078-0432.CCR-19-3545
    1. Schmid P, Adams S, Rugo HS, et al. . Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N Engl J Med 2018;379:2108–21. 10.1056/NEJMoa1809615
    1. Rugo HS, Delord J-P, Im S-A, et al. . Safety and antitumor activity of pembrolizumab in patients with estrogen Receptor-Positive/Human epidermal growth factor receptor 2-Negative advanced breast cancer. Clin Cancer Res 2018;24:2804–11. 10.1158/1078-0432.CCR-17-3452
    1. Loi S, Sirtaine N, Piette F, et al. . Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin with doxorubicin-based chemotherapy: big 02-98. J Clin Oncol 2013;31:860–7. 10.1200/JCO.2011.41.0902
    1. Patel SP, Kurzrock R. Pd-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther 2015;14:847–56. 10.1158/1535-7163.MCT-14-0983
    1. Davis AA, Patel VG. The role of PD-L1 expression as a predictive biomarker: an analysis of all US food and drug administration (FDA) approvals of immune checkpoint inhibitors. J Immunother Cancer 2019;7:278. 10.1186/s40425-019-0768-9
    1. Spring LM, Wander SA, Zangardi M, et al. . CDK 4/6 inhibitors in breast cancer: current controversies and future directions. Curr Oncol Rep 2019;21:25. 10.1007/s11912-019-0769-3
    1. Finn RS, Martin M, Rugo HS, et al. . Palbociclib and letrozole in advanced breast cancer. N Engl J Med 2016;375:1925–36. 10.1056/NEJMoa1607303
    1. Deng J, Wang ES, Jenkins RW, et al. . CDK4/6 inhibition augments antitumor immunity by enhancing T-cell activation. Cancer Discov 2018;8:216–33. 10.1158/-17-0915
    1. Teo ZL, Versaci S, Dushyanthen S, et al. . Combined CDK4/6 and PI3Kα inhibition is synergistic and immunogenic in triple-negative breast cancer. Cancer Res 2017;77:6340–52. 10.1158/0008-5472.CAN-17-2210
    1. Goel S, DeCristo MJ, Watt AC, et al. . CDK4/6 inhibition triggers anti-tumour immunity. Nature 2017;548:471–5. 10.1038/nature23465
    1. Schaer DA, Beckmann RP, Dempsey JA, et al. . The CDK4/6 inhibitor Abemaciclib induces a T cell inflamed tumor microenvironment and enhances the efficacy of PD-L1 checkpoint blockade. Cell Rep 2018;22:2978–94. 10.1016/j.celrep.2018.02.053
    1. Yuan Y, Yost SE, Lee JS. Abstract P3-11-04: a phase II study of pembrolizumab, letrozole and palbociclib in patients with metastatic estrogen receptor positive breast cancer. Cancer Research 2020.
    1. Salgado R, Denkert C, Demaria S, et al. . The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an international TILs Working group 2014. Ann Oncol 2015;26:259–71. 10.1093/annonc/mdu450
    1. Chevrier S, Crowell HL, Zanotelli VRT, et al. . Compensation of signal spillover in suspension and imaging mass cytometry. Cell Syst 2018;6:612–20. 10.1016/j.cels.2018.02.010
    1. Beaubier N, Tell R, Lau D, et al. . Clinical validation of the tempus xT next-generation targeted oncology sequencing assay. Oncotarget 2019;10:2384–96. 10.18632/oncotarget.26797
    1. Newman AM, Liu CL, Green MR, et al. . Robust enumeration of cell subsets from tissue expression profiles. Nat Methods 2015;12:453–7. 10.1038/nmeth.3337
    1. Sade-Feldman M, Yizhak K, Bjorgaard SL, et al. . Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 2018;175:998–1013. 10.1016/j.cell.2018.10.038
    1. Im SJ, Hashimoto M, Gerner MY, et al. . Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 2016;537:417–21. 10.1038/nature19330
    1. Thommen DS, Koelzer VH, Herzig P, et al. . A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat Med 2018;24:994–1004. 10.1038/s41591-018-0057-z
    1. Bronte V, Brandau S, Chen S-H, et al. . Recommendations for myeloid-derived suppressor cell Nomenclature and characterization standards. Nat Commun 2016;7:12150. 10.1038/ncomms12150
    1. Havel JJ, Chowell D, Chan TA. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat Rev Cancer 2019;19:133–50. 10.1038/s41568-019-0116-x
    1. Manjarrez-Orduño N, Menard LC, Kansal S, et al. . Circulating T cell subpopulations correlate with immune responses at the tumor site and clinical response to PD1 inhibition in non-small cell lung cancer. Front Immunol 2018;9:1613. 10.3389/fimmu.2018.01613
    1. Tietze JK, Angelova D, Heppt MV, et al. . The proportion of circulating CD45RO+CD8+ memory T cells is correlated with clinical response in melanoma patients treated with ipilimumab. Eur J Cancer 2017;75:268–79. 10.1016/j.ejca.2016.12.031
    1. Kunert A, Basak EA, Hurkmans DP, et al. . CD45RA+CCR7- CD8 T cells lacking co-stimulatory receptors demonstrate enhanced frequency in peripheral blood of NSCLC patients responding to nivolumab. J Immunother Cancer 2019;7:149. 10.1186/s40425-019-0608-y
    1. Sathaliyawala T, Kubota M, Yudanin N, et al. . Distribution and compartmentalization of human circulating and tissue-resident memory T cell subsets. Immunity 2013;38:187–97. 10.1016/j.immuni.2012.09.020
    1. Joshi NS, Cui W, Chandele A, et al. . Inflammation directs memory precursor and short-lived effector CD8(+) T cell fates via the graded expression of T-bet transcription factor. Immunity 2007;27:281–95. 10.1016/j.immuni.2007.07.010
    1. Herndler-Brandstetter D, Ishigame H, Shinnakasu R, et al. . KLRG1+ Effector CD8+ T Cells Lose KLRG1, Differentiate into All Memory T Cell Lineages, and Convey Enhanced Protective Immunity. Immunity 2018;48:716–29. 10.1016/j.immuni.2018.03.015
    1. Wei SC, Levine JH, Cogdill AP, et al. . Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade. Cell 2017;170:1120–33. 10.1016/j.cell.2017.07.024
    1. Liakou CI, Kamat A, Tang DN, et al. . CTLA-4 blockade increases IFNgamma-producing CD4+ICOShi cells to shift the ratio of effector to regulatory T cells in cancer patients. Proc Natl Acad Sci U S A 2008;105:14987–92. 10.1073/pnas.0806075105
    1. Ng Tang D, Shen Y, Sun J, et al. . Increased frequency of ICOS+ CD4 T cells as a pharmacodynamic biomarker for anti-CTLA-4 therapy. Cancer Immunol Res 2013;1:229–34. 10.1158/2326-6066.CIR-13-0020
    1. Kamphorst AO, Pillai RN, Yang S, et al. . Proliferation of PD-1+ CD8 T cells in peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc Natl Acad Sci U S A 2017;114:4993–8. 10.1073/pnas.1705327114
    1. Huang AC, Orlowski RJ, Xu X, et al. . A single dose of neoadjuvant PD-1 blockade predicts clinical outcomes in resectable melanoma. Nat Med 2019;25:454–61. 10.1038/s41591-019-0357-y
    1. Williford J-M, Ishihara J, Ishihara A, et al. . Recruitment of CD103+ dendritic cells via tumor-targeted chemokine delivery enhances efficacy of checkpoint inhibitor immunotherapy. Sci Adv 2019;5:eaay1357. 10.1126/sciadv.aay1357
    1. Molgora M, Esaulova E, Vermi W, et al. . Trem2 modulation remodels the tumor myeloid landscape enhancing anti-PD-1 immunotherapy. Cell 2020;182:886–900. 10.1016/j.cell.2020.07.013
    1. Bergenfelz C, Larsson A-M, von Stedingk K, et al. . Systemic monocytic-MDSCs are generated from monocytes and correlate with disease progression in breast cancer patients. PLoS One 2015;10:e0127028. 10.1371/journal.pone.0127028
    1. Wang Q, Guldner IH, Golomb SM, et al. . Single-cell profiling guided combinatorial immunotherapy for fast-evolving CDK4/6 inhibitor-resistant HER2-positive breast cancer. Nat Commun 2019;10:1–12. 10.1038/s41467-019-11729-1
    1. Xue J, Schmidt SV, Sander J, et al. . Transcriptome-Based network analysis reveals a spectrum model of human macrophage activation. Immunity 2014;40:274–88. 10.1016/j.immuni.2014.01.006
    1. Murray PJ. Macrophage polarization. Annu Rev Physiol 2017;79:541–66. 10.1146/annurev-physiol-022516-034339
    1. Cassetta L, Fragkogianni S, Sims AH, et al. . Human tumor-associated macrophage and monocyte transcriptional landscapes reveal cancer-specific reprogramming, biomarkers, and therapeutic targets. Cancer Cell 2019;35:588–602. 10.1016/j.ccell.2019.02.009
    1. Kitamura T, Doughty-Shenton D, Cassetta L, et al. . Monocytes differentiate to immune suppressive precursors of metastasis-associated macrophages in mouse models of metastatic breast cancer. Front Immunol 2017;8:2004. 10.3389/fimmu.2017.02004
    1. Spranger S, Dai D, Horton B, et al. . Tumor-Residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell 2017;31:711–23. 10.1016/j.ccell.2017.04.003
    1. Garris CS, Arlauckas SP, Kohler RH, et al. . Successful anti-PD-1 cancer immunotherapy requires T Cell-Dendritic cell crosstalk involving the cytokines IFN-γ and IL-12. Immunity 2018;49:1148–61. 10.1016/j.immuni.2018.09.024
    1. SA O, D-C W, Cheung J. PD-L1 expression by dendritic cells is a key regulator of T-cell immunity in cancer. Nat Cancer 2020;1:681–91. 10.1038/s43018-020-0075-x

Source: PubMed

3
Subscribe