First-in-Class Anti-immunoglobulin-like Transcript 4 Myeloid-Specific Antibody MK-4830 Abrogates a PD-1 Resistance Mechanism in Patients with Advanced Solid Tumors

Lillian L Siu, Ding Wang, John Hilton, Ravit Geva, Drew Rasco, Ruth Perets, Anson K Abraham, Douglas C Wilson, Julia F Markensohn, Jared Lunceford, Leah Suttner, Shabana Siddiqi, Rachel A Altura, Corinne Maurice-Dror, Lillian L Siu, Ding Wang, John Hilton, Ravit Geva, Drew Rasco, Ruth Perets, Anson K Abraham, Douglas C Wilson, Julia F Markensohn, Jared Lunceford, Leah Suttner, Shabana Siddiqi, Rachel A Altura, Corinne Maurice-Dror

Abstract

Purpose: In this first-in-human study (NCT03564691) in advanced solid tumors, we investigated a novel first-in-class human IgG4 monoclonal antibody targeting the immunoglobulin-like transcript 4 (ILT4) receptor, MK-4830, as monotherapy and in combination with pembrolizumab.

Patients and methods: Patients with histologically/cytologically confirmed advanced solid tumors, measurable disease by RECIST v1.1, and evaluable baseline tumor sample received escalating doses of intravenous MK-4830 every 3 weeks as monotherapy (parts A and B) and in combination with pembrolizumab (part C). Safety and tolerability were the primary objectives. Pharmacokinetics, objective response rate per RECIST v1.1, and molecular biomarkers were also evaluated.

Results: Of 84 patients, 50 received monotherapy and 34 received combination therapy. No dose-limiting toxicities were observed; maximum tolerated dose was not reached. MK-4830 showed dose-related target engagement. Eleven of 34 patients in the dose-escalation phase who received combination therapy achieved objective responses; 5 previously had progressive disease on anti-PD-1/PD-L1 therapies. Exploratory evaluation of the association between response and pretreatment gene expression related to interferon-gamma signaling in tumors suggested higher sensitivity to T-cell inflammation with combination therapy than historically expected with pembrolizumab monotherapy, with greater response at more moderate levels of inflammation.

Conclusions: This first-in-class MK-4830 antibody dosed as monotherapy and in combination with pembrolizumab was well tolerated with no unexpected toxicities, and demonstrated dose-related evidence of target engagement and antitumor activity. Inflammation intrinsic to the ILT4 mechanism may be facilitated by alleviating the myeloid-suppressive components of the tumor microenvironment, supporting the target of ILT4 as a potential novel immunotherapy in combination with an anti-PD-1/PD-L1 agent.

©2021 The Authors; Published by the American Association for Cancer Research.

Figures

Figure 1.
Figure 1.
Proposed mechanism of action of MK-4830. ILT4, immunoglobulin-like transcript 4; PD-1, programmed death 1.
Figure 2.
Figure 2.
Target lesion change over time (RECIST v1.1) in patients who received (A) MK-4830 monotherapy or (B) MK-4830 in combination with pembrolizumab. Best percentage change from baseline in target lesion based on investigator assessment (RECIST v1.1) in patients who received (C) MK-4830 monotherapy or (D) MK-4830 in combination with pembrolizumab. RECIST v1.1, Response Evaluation Criteria in Solid Tumors version 1.1.
Figure 3.
Figure 3.
Association between response and (A) PD-L1 status, (B) TMB status, and (C) TcellinfGEP score. AUROC, area under the receiver operating characteristic; CI, confidence interval; CPS, combined positive score; CR, complete response; NR, nonresponder; PD, progressive disease; PD-L1, programmed death ligand 1; PR, partial response; R, responder; SD, stable disease; TcellinfGEP, T-cell–inflamed gene expression profile; TMB, tumor mutational burden.
Figure 4.
Figure 4.
Correlation between myeloid-specific biomarkers and (A) PD-L1 status, (B) TMB status, (C) TcellinfGEP score, and (D) other myeloid-specific biomarkers. CPS, combined positive score; CR, complete response; HLA-G, human leukocyte antigen G; LILRB2, leukocyte immunoglobulin-like receptor B2; mMDSC, monocytic myeloid-derived suppressor cell; NR, nonresponder; PD, progressive disease; PD-L1, programmed death ligand 1; PR, partial response; R, responder; SD, stable disease; TcellinfGEP, T-cell–inflamed gene expression profile; TMB, tumor mutational burden.
Figure 5.
Figure 5.
Association between response and TcellinfGEP-adjusted (A) LILRB2, (B) mMDSC, and (C) HLA-G. AUROC, area under the receiver operating characteristic; CI, confidence interval; CR, complete response; HLA-G, human leukocyte antigen G; LILRB2, leukocyte immunoglobulin-like receptor B2; mMDSC, monocytic myeloid-derived suppressor cell; NR, nonresponder; PD, progressive disease; PR, partial response; R, responder; SD, stable disease; TcellinfGEP, T-cell–inflamed gene expression profile.

References

    1. Pitt JM, Vétizou M, Daillère R, Roberti MP, Yamazaki T, Routy B, et al. . Resistance mechanisms to immune-checkpoint blockade in cancer: tumor-intrinsic and -extrinsic factors. Immunity 2016;44:1255–69.
    1. Rouas-Freiss N, LeMaoult J, Verine J, Tronik-Le Roux D, Culine S, Hennequin C, et al. . Intratumor heterogeneity of immune checkpoints in primary renal cell cancer: focus on HLA-G/ILT2/ILT4. Oncoimmunology 2017;6:e1342023.
    1. Hirano F, Kaneko K, Tamura H, Dong H, Wang S, Ichikawa M, et al. . Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res 2005;65:1089–96.
    1. Francisco LM, Salinas VH, Brown KE, Vanguri VK, Freeman GJ, Kuchroo VK, et al. . PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J Exp Med 2009;206:3015–29.
    1. Fleming V, Hu X, Weber R, Nagibin V, Groth C, Altevogt P, et al. . Targeting myeloid-derived suppressor cells to bypass tumor-induced immunosuppression. Front Immunol 2018;9:398.
    1. Ai L, Mu S, Wang Y, Wang H, Cai L, Li W, et al. . Prognostic role of myeloid-derived suppressor cells in cancers: a systematic review and meta-analysis. BMC Cancer 2018;18:1220.
    1. Adah D, Hussain M, Qin L, Qin L, Zhang J, Chen X. Implications of MDSCs-targeting in lung cancer chemo-immunotherapeutics. Pharmacol Res 2016;110:25–34.
    1. De Cicco P, Ercolano G, Ianaro A. The new era of cancer immunotherapy: targeting myeloid-derived suppressor cells to overcome immune evasion. Front Immunol 2020;11:1680.
    1. Colonna M, Navarro F, Bellón T, Llano M, García P, Samaridis J, et al. . A common inhibitory receptor for major histocompatibility complex class I molecules on human lymphoid and myelomonocytic cells. J Exp Med 1997;186:1809–18.
    1. Colonna M, Samaridis J, Cella M, Angman L, Allen RL, O'Callaghan CA, et al. . Human myelomonocytic cells express an inhibitory receptor for classical and nonclassical MHC class I molecules. J Immunol 1998;160:3096–100.
    1. Fanger NA, Cosman D, Peterson L, Braddy SC, Maliszewski CR, Borges L. The MHC class I binding proteins LIR-1 and LIR-2 inhibit Fc receptor-mediated signaling in monocytes. Eur J Immunol 1998;28:3423–34.
    1. Gao A, Sun Y, Peng G. ILT4 functions as a potential checkpoint molecule for tumor immunotherapy. Biochim Biophys Acta Rev Cancer 2018;1869:278–85.
    1. Köstlin N, Ostermeir AL, Spring B, Schwarz J, Marmé A, Walter CB, et al. . HLA-G promotes myeloid-derived suppressor cell accumulation and suppressive activity during human pregnancy through engagement of the receptor ILT4. Eur J Immunol 2017;47:374–84.
    1. Cai Z, Wang L, Han Y, Gao W, Wei X, Gong R, et al. . Immunoglobulin-like transcript 4 and human leukocyte antigen-G interaction promotes the progression of human colorectal cancer. Int J Oncol 2019;54:1943–54.
    1. Li Q, Li J, Wang S, Wang J, Chen X, Zhou D, et al. . Overexpressed immunoglobulin-like transcript (ILT) 4 in lung adenocarcinoma is correlated with immunosuppressive T-cell subset infiltration and poor patient outcomes. Biomark Res 2020;8:11.
    1. Chen HM, van der Touw W, Wang YS, Kang K, Mai S, Zhang J, et al. . Blocking immunoinhibitory receptor LILRB2 reprograms tumor-associated myeloid cells and promotes antitumor immunity. J Clin Invest 2018;128:5647–62.
    1. Sharma MD, Rodriguez PC, Koehn BH, Baban B, Cui Y, Guo G, et al. . Activation of p53 in immature myeloid precursor cells controls differentiation into Ly6c(+)CD103(+) monocytic antigen-presenting cells in tumors. Immunity 2018;48:91–106.
    1. Zuniga L, Joyce-Shaikh B, Wilson D, Cherwinski H, Chen Y, Jeff G, et al. . Preclinical characterization of a first-in-class ILT4 antagonist, MK-4830. J ImmunoTher Can 2018;6:115.
    1. KEYTRUDA® (pembrolizumab) injection, for intravenous use. Merck Sharp & Dohme Corp.: Whitehouse Station, NJ, USA; 2021.
    1. Ayers M, Lunceford J, Nebozhyn M, Murphy E, Loboda A, Kaufman DR, et al. . IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest 2017;127:2930–40.
    1. Cristescu R, Nebozhyn M, Zhang C, Albright A, Kobie J, Huang L, et al. . Pan-tumor analysis of the association of cancer and immune biology-related gene expression signatures with response to pembrolizumab monotherapy. J ImmunoTher Can 2019;7:282.
    1. Seymour L, Bogaerts J, Perrone A, Ford R, Schwartz LH, Mandrekar S, et al. . iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol 2017;18:e143–e52.
    1. Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, et al. . Pembrolizumab for the treatment of non–small cell lung cancer. N Engl J Med 2015;372:2018–28.
    1. Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, et al. . Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med 2015;372:2521–32.
    1. van Vugt MJH, Stone JA, De Greef R, Snyder ES, Lipka L, Turner DC, et al. . Immunogenicity of pembrolizumab in patients with advanced tumors. J Immunother Can 2019;7:212.
    1. Cristescu R, Mogg R, Ayers M, Albright A, Murphy E, Yearley J, et al. . Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science 2018;362:eaar3593.

Source: PubMed

3
Abonneren