CDK4/6 inhibition enhances antitumor efficacy of chemotherapy and immune checkpoint inhibitor combinations in preclinical models and enhances T-cell activation in patients with SCLC receiving chemotherapy

Anne Y Lai, Jessica A Sorrentino, Konstantin H Dragnev, Jared M Weiss, Taofeek K Owonikoko, Julie A Rytlewski, Jill Hood, Zhao Yang, Rajesh K Malik, Jay C Strum, Patrick J Roberts, Anne Y Lai, Jessica A Sorrentino, Konstantin H Dragnev, Jared M Weiss, Taofeek K Owonikoko, Julie A Rytlewski, Jill Hood, Zhao Yang, Rajesh K Malik, Jay C Strum, Patrick J Roberts

Abstract

Background: Combination treatment with chemotherapy and immune checkpoint inhibitors (ICIs) has demonstrated meaningful clinical benefit to patients. However, chemotherapy-induced damage to the immune system can potentially diminish the efficacy of chemotherapy/ICI combinations. Trilaciclib, a highly potent, selective and reversible cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitor in development to preserve hematopoietic stem and progenitor cells and immune system function during chemotherapy, has demonstrated proof of concept in recent clinical trials. Furthermore, CDK4/6 inhibition has been shown to augment T-cell activation and antitumor immunity in preclinical settings. Therefore, addition of trilaciclib has the potential to further enhance the efficacy of chemotherapy and ICI combinations.

Methods: In murine syngeneic tumor models, a schedule of 3 weekly doses of trilaciclib was combined with chemotherapy/ICI regimens to assess the effect of transient CDK4/6 inhibition on antitumor response and intratumor T-cell proliferation and function. Peripheral T-cell status was also analyzed in patients with small cell lung cancer (SCLC) treated with chemotherapy with or without trilaciclib to gain insights into the effect of transient exposure of trilaciclib on T-cell activation.

Results: Preclinically, the addition of trilaciclib to chemotherapy/ICI regimens enhanced antitumor response and overall survival compared with chemotherapy and ICI combinations alone. This effect is associated with the modulation of the proliferation and composition of T-cell subsets in the tumor microenvironment and increased effector function. Transient exposure of trilaciclib in patients with SCLC during chemotherapy treatment both preserved and increased peripheral lymphocyte counts and enhanced T-cell activation, suggesting that trilaciclib not only preserved but also enhanced immune system function.

Conclusions: Transient CDK4/6 inhibition by trilaciclib was sufficient to enhance and prolong the duration of the antitumor response by chemotherapy/ICI combinations, suggesting a role for the transient cell cycle arrest of tumor immune infiltrates in remodeling the tumor microenvironment. These results provide a rationale for combining trilaciclib with chemotherapy/ICI regimens to improve antitumor efficacy in patients with cancer.

Trial registration: ClinicalTrials.gov NCT02499770.

Keywords: clinical trials; drug evaluation; investigational; phase II as topic; preclinical; therapies.

Conflict of interest statement

Competing interests: AYL, PJR, ZY and JCS were employees of G1 Therapeutics at the time of the study. AYL is an employee of Locus Biosciences. PJR and JCS are employees of Arc Therapeutics. ZY is an employee of Everest Clinical Research. JAS and RKM are employees of G1 Therapeutics. JAR was an employee of Adaptive Biotechnologies at the time of the study. JH is an employee of and has a financial interest in Adaptive Biotechnologies. JMW has received honorarium from G1 Therapeutics. JMW, KHD and TKO received research funding from G1 Therapeutics (to their institution).

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Addition of trilaciclib to oxaliplatin and αPD-L1 combination therapy, and other multiple chemotherapy and ICI combinations, led to enhanced antitumor activity and durability of response in MC38 and CT26 tumor models. (A) Three dosing schedules were initially tested in the MC38 model. The induction plus maintenance schedule was carried forward in all other experiments in both the MC38 and CT26 models with the substitution of 5-FU for oxaliplatin, or anti-PD-1 for anti-PD-L1 as indicated. (B–D) Tumor growth and overall survival of MC38 and CT26 mice treated with chemotherapy/ICI ±trilaciclib combination therapy (n=10–15 per treatment group). *P≤0.05. 5-FU, 5-fluorouracil; αPD-L1, anti-programmed death-ligand-1; I, induction; ICI, immune checkpoint inhibitor; IM, induction plus maintenance; M, maintenance; OP, oxaliplatin plus anti-programmed death-ligand-1; TOP, trilaciclib plus oxaliplatin and anti-programmed death-ligand-1.
Figure 2
Figure 2
Addition of trilaciclib to OP treatment combination resulted in transient proliferation arrest followed by a faster recovery of CD8+ and CD4+ T cells compared with Tregs. (A) Baseline percent proliferation status of immune cell populations in spleen and tumors in MC38 tumor-bearing mice (n=16 biological replicates), and proliferation of intratumor lymphoid and myeloid immune cell types at (B) 6–24 hours (n=4 biological replicates) and (C) days 2, 4 and 7 (n=4 or 5 biological replicates) after trilaciclib treatment. Percent proliferation was defined as proportion of EdU+ cells, and relative proliferation was defined as (% EdU+ cells in trilaciclib-treated samples)/(% EdU+ cells in vehicle-treated samples) x 100. Each biological replicate consists of a pool of 3 animals. (D) Proliferation of CD8+ T cells in coculture with trilaciclib-treated Tregs (n=3 independent experiments, each performed with three biological replicates per culture condition). Data represent mean±SD. *p<0.05; **p<0.01. EdU, 5-ethynyl-2ʼ-deoxyuridine; gMDSC, granulocytic myeloid-derived suppressor cell; mMDSC, monocytic myeloid-derived suppressor cell; NK, natural killer; OP, oxaliplatin plus anti-programmed death-ligand-1; TOP, trilaciclib plus oxaliplatin and anti-programmed death-ligand-1; Treg, T-regulatory cell.
Figure 3
Figure 3
Transient G1 arrest led to changes in the proportion of intratumor T-cell subsets favoring effector T-cell function. (A) Flow cytometric analysis of intratumor CD8+ T cells, CD4+ T cells, myeloid cell types (macrophages, mMDSC and gMDSC populations) and Tregs; (B) proportion of Treg in total tumor or spleen CD4+ T cells and (C) ratio of CD8+ T cells to Tregs (% CD8+ T cells/% Tregs) in the CD45+ population or spleen (n=5–8 tumors analyzed per treatment group and time point); and (D) proportion of activated (% CD69+) cells in CD8+ or CD4+ T cells. *P<0.05. gMDSC, granulocytic myeloid-derived suppressor cell; mMDSC, monocytic myeloid-derived suppressor cell; OP, oxaliplatin plus anti-programmed death-ligand-1; TOP, trilaciclib plus oxaliplatin and anti-programmed death-ligand-1; Treg, T-regulatory cell.
Figure 4
Figure 4
Trilaciclib in combination with chemotherapy and ICI enhanced the gene signature of cytotoxic antitumor response. (A) Between-group comparison of the geometric mean of log2-normalized transcript levels of genes upregulated in either OP or TOP versus vehicle associated with each KEGG pathway and (B) associated heat maps showing mean log2 transcript levels of each gene in the pathway; and (C) T-cell repertoire analyses of MC38 tumors from OP-treated or TOP-treated mice (n=5 per treatment group) on day 17, presented as either clonality score and proportion of T cells in tumor (T-cell fraction; left panel), or median clonality score and TIL fraction (right panel). Error bars denote SE, and dotted lines represent the median value for TIL fraction and clonality score. *P

Figure 5

Addition of trilaciclib to E/P…

Figure 5

Addition of trilaciclib to E/P preserved and enhanced lymphocyte counts and function in…

Figure 5
Addition of trilaciclib to E/P preserved and enhanced lymphocyte counts and function in patients with SCLC. (A) Mean absolute cell count (cells/µL) for B-cell and T-cell subsets and activated CD8+ T cells (CD3+CD8+CD38+HLA-DR+), and ratio of absolute whole blood cell counts for CD8+ T cells to Tregs (CD3+CD4+CD25+CD127low), at the indicated time points. Error bars represent 95% CIs. (B) Per-patient T-cell repertoire analysis comparing mean ratio of clonality versus pretreatment level (C1D1) and number of expanded T-cell clones at the indicated time points. Bars represent median and IQR. (C) Proportion of patients with high (≥median) number of expanded T-cell clones at C3D1 (≥15 clones) and C5D1 (≥17 clones). (D) Progression-free and overall survival analysis of all patients with low (<median) and high (≥median) numbers of expanded T-cell clones on C3D1 and C5D1. *P=0.0098. C1D1, cycle 1 day 1; C3D1, cycle 3 day 1; C5D1, cycle 5 day 1; D, day; E/P, etoposide and carboplatin; T/E/P, trilaciclib prior to etoposide and carboplatin; PTV, post-treatment visit; SCLC, small cell lung cancer; Treg, T-regulatory cell.
Figure 5
Figure 5
Addition of trilaciclib to E/P preserved and enhanced lymphocyte counts and function in patients with SCLC. (A) Mean absolute cell count (cells/µL) for B-cell and T-cell subsets and activated CD8+ T cells (CD3+CD8+CD38+HLA-DR+), and ratio of absolute whole blood cell counts for CD8+ T cells to Tregs (CD3+CD4+CD25+CD127low), at the indicated time points. Error bars represent 95% CIs. (B) Per-patient T-cell repertoire analysis comparing mean ratio of clonality versus pretreatment level (C1D1) and number of expanded T-cell clones at the indicated time points. Bars represent median and IQR. (C) Proportion of patients with high (≥median) number of expanded T-cell clones at C3D1 (≥15 clones) and C5D1 (≥17 clones). (D) Progression-free and overall survival analysis of all patients with low (<median) and high (≥median) numbers of expanded T-cell clones on C3D1 and C5D1. *P=0.0098. C1D1, cycle 1 day 1; C3D1, cycle 3 day 1; C5D1, cycle 5 day 1; D, day; E/P, etoposide and carboplatin; T/E/P, trilaciclib prior to etoposide and carboplatin; PTV, post-treatment visit; SCLC, small cell lung cancer; Treg, T-regulatory cell.

References

    1. Bisi JE, Sorrentino JA, Roberts PJ, et al. . Preclinical characterization of G1T28: a novel CDK4/6 inhibitor for reduction of chemotherapy-induced myelosuppression. Mol Cancer Ther 2016;15:783–93. 10.1158/1535-7163.MCT-15-0775
    1. He S, Roberts PJ, Sorrentino JA, et al. . Transient CDK4/6 inhibition protects hematopoietic stem cells from chemotherapy-induced exhaustion. Sci Transl Med 2017;9:eaal3986 10.1126/scitranslmed.aal3986
    1. Weiss JM, Csoszi T, Maglakelidze M, et al. . Myelopreservation with the CDK4/6 inhibitor trilaciclib in patients with small-cell lung cancer receiving first-line chemotherapy: a phase Ib/randomized phase II trial. Ann Oncol 2019;30:1613–21. 10.1093/annonc/mdz278
    1. Tan AR, Wright GS, Thummala AR, et al. . Trilaciclib plus chemotherapy versus chemotherapy alone in patients with metastatic triple-negative breast cancer: a multicentre, randomised, open-label, phase 2 trial. Lancet Oncol 2019;20:1587–601. 10.1016/S1470-2045(19)30616-3
    1. Klein ME, Kovatcheva M, Davis LE, et al. . CDK4/6 inhibitors: the mechanism of action may not be as simple as once thought. Cancer Cell 2018;34:9–20. 10.1016/j.ccell.2018.03.023
    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. 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. Zhang J, Bu X, Wang H, et al. . Cyclin D-CDK4 kinase destabilizes PD-L1 via cullin 3-SPOP to control cancer immune surveillance. Nature 2018;553:91–5. 10.1038/nature25015
    1. Jerby-Arnon L, Shah P, Cuoco MS, et al. . A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell 2018;175:e24:984–97. 10.1016/j.cell.2018.09.006
    1. Reck M, Rodríguez-Abreu D, Robinson AG, et al. . Pembrolizumab versus chemotherapy for PD-L1–positive non-small-cell lung cancer. N Engl J Med 2016;375:1823–33. 10.1056/NEJMoa1606774
    1. Gandhi L, Rodríguez-Abreu D, Gadgeel S, et al. . Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med 2018;378:2078–92. 10.1056/NEJMoa1801005
    1. Paz-Ares L, Luft A, Vicente D, et al. . Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer. N Engl J Med 2018;379:2040–51. 10.1056/NEJMoa1810865
    1. Horn L, Mansfield AS, Szczęsna A, et al. . First-Line atezolizumab plus chemotherapy in extensive-stage small-cell lung cancer. N Engl J Med 2018;379:2220–9. 10.1056/NEJMoa1809064
    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. Pfirschke C, Engblom C, Rickelt S, et al. . Immunogenic chemotherapy sensitizes tumors to checkpoint blockade therapy. Immunity 2016;44:343–54. 10.1016/j.immuni.2015.11.024
    1. North RJ. Cyclophosphamide-facilitated adoptive immunotherapy of an established tumor depends on elimination of tumor-induced suppressor T cells. J Exp Med 1982;155:1063–74. 10.1084/jem.155.4.1063
    1. Suzuki E, Kapoor V, Jassar AS, et al. . Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clin Cancer Res 2005;11:6713–21. 10.1158/1078-0432.CCR-05-0883
    1. Zitvogel L, Apetoh L, Ghiringhelli F, et al. . Immunological aspects of cancer chemotherapy. Nat Rev Immunol 2008;8:59–73. 10.1038/nri2216
    1. Smith RE. Trends in recommendations for myelosuppressive chemotherapy for the treatment of solid tumors. J Natl Compr Canc Netw 2006;4:649–58. 10.6004/jnccn.2006.0056
    1. Lyman GH. Risks and consequences of chemotherapy-induced neutropenia. Clin Cornerstone 2006;8 Suppl 5:S12–18. 10.1016/S1098-3597(06)80054-2
    1. Corbett TH, Griswold DP, Roberts BJ, et al. . Tumor induction relationships in development of transplantable cancers of the colon in mice for chemotherapy assays, with a note on carcinogen structure. Cancer Res 1975;35:2434–9.
    1. Bates D, Machler M, Bolker B, et al. . Fitting linear mixed-effects models using lme4. J Stat Softw 2015;67:1–48. Available from:
    1. Kuznetsova A, Brockhoff PB, Christensen RHB. lmerTest package: tests in linear mixed effects models. J Stat Softw 2017;82:1–26. 10.18637/jss.v082.i13
    1. Robins HS, Campregher PV, Srivastava SK, et al. . Comprehensive assessment of T-cell receptor β-chain diversity in αβ T cells. Blood 2009;114:4099–107. 10.1182/blood-2009-04-217604
    1. Wu D, Emerson RO, Sherwood A, et al. . Detection of minimal residual disease in B lymphoblastic leukemia by high-throughput sequencing of IgH. Clin Cancer Res 2014;20:4540–8. 10.1158/1078-0432.CCR-13-3231
    1. Tumeh PC, Harview CL, Yearley JH, et al. . PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014;515:568–71. 10.1038/nature13954
    1. Hsu M, Sedighim S, Wang T, et al. . TCR sequencing can identify and track glioma-infiltrating T cells after DC vaccination. Cancer Immunol Res 2016;4:412–8. 10.1158/2326-6066.CIR-15-0240
    1. DeWitt WS, Emerson RO, Lindau P, et al. . Dynamics of the cytotoxic T cell response to a model of acute viral infection. J Virol 2015;89:4517–26. 10.1128/JVI.03474-14
    1. Tiessen RG, Roberts PJ, Sorrentino JA, et al. . First-in-human phase 1 safety, PK, and PD study of the CDK4/6 inhibitor G1T28. J Clin Oncol 2015;33:2527.10.1200/jco.2015.33.15_suppl.2527
    1. Roberts PJ, White HS, Sorrentino JA, et al. . Evaluation of targeted bone marrow arrest by G1T28, a CDK4/6 inhibitor in clinical development to reduce chemotherapy-induced myelosuppression. J Clin Oncol 2015;33:2529.10.1200/jco.2015.33.15_suppl.2529
    1. Ayers M, Lunceford J, Nebozhyn M, et al. . IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest 2017;127:2930–40. 10.1172/JCI91190
    1. Amaria RN, Reddy SM, Tawbi HA, et al. . Neoadjuvant immune checkpoint blockade in high-risk resectable melanoma. Nat Med 2018;24:1649–54. 10.1038/s41591-018-0197-1
    1. Roberts P, He S, Schvartsman G, et al. . G1T28, a CDK4/6 inhibitor, preserves T lymphocyte function from damage by cytotoxic chemotherapy. Eur J Cancer 2016;69:S143–4. 10.1016/S0959-8049(16)33026-X
    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. Ku GY, Yuan J, Page DB, et al. . Single-institution experience with ipilimumab in advanced melanoma patients in the compassionate use setting: lymphocyte count after 2 doses correlates with survival. Cancer 2010;116:1767–75. 10.1002/cncr.24951
    1. Huang AC, Postow MA, Orlowski RJ, et al. . T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 2017;545:60–5. 10.1038/nature22079
    1. George J, Lim JS, Jang SJ, et al. . Comprehensive genomic profiles of small cell lung cancer. Nature 2015;524:47–53. 10.1038/nature14664
    1. Wu TD, Madireddi S, de Almeida PE, et al. . Peripheral T cell expansion predicts tumour infiltration and clinical response. Nature 2020;579:274–8. 10.1038/s41586-020-2056-8
    1. Semenova EA, Nagel R, Berns A. Origins, genetic landscape, and emerging therapies of small cell lung cancer. Genes Dev 2015;29:1447–62. 10.1101/gad.263145.115
    1. Pavan A, Attili I, Pasello G, et al. . Immunotherapy in small-cell lung cancer: from molecular promises to clinical challenges. J Immunother Cancer 2019;7:205. 10.1186/s40425-019-0690-1
    1. Daniel D, Kuchava V, Bondarenko I, et al. . Trilaciclib (T) decreases myelosuppression in extensive-stage small cell lung cancer (ES-SCLC) patients receiving first-line chemotherapy plus atezolizumab. Ann Oncol 2019;30:v710–7. 10.1093/annonc/mdz264.006
    1. Xiao Y, Ma D, Zhao S, et al. . Multi-omics profiling reveals distinct microenvironment characterization and suggests immune escape mechanisms of triple-negative breast cancer. Clin Cancer Res 2019;25:5002–14. 10.1158/1078-0432.CCR-18-3524

Source: PubMed

3
订阅