Elsevier

Cytotherapy

Volume 22, Issue 4, April 2020, Pages 204-213
Cytotherapy

FULL-LENGTH ARTICLE
Clinical Studies
Common phenotypic dynamics of tumor-infiltrating lymphocytes across different histologies upon checkpoint inhibition: impact on clinical outcome

https://doi.org/10.1016/j.jcyt.2020.01.010Get rights and content

Abstract

Background

Immune checkpoint inhibitors (ICIs) have revolutionized the cancer therapeutic landscape and our perception of interactions between the immune system and tumor cells. Despite remarkable progress, disease relapse and primary resistance are not uncommon. Understanding the biological processes that tumor-infiltrating lymphocytes (TILs) undergo during ICI, how this affects the tumor microenvironment (TME) and, ultimately, clinical outcome is, therefore, necessary to further improve treatment efficacy.

Aim

In the current study, we sought to characterize TILs from patients with metastatic solid tumors undergoing ICI correlating flowcytometric findings with clinical outcome.

Methods

In total, 20 patients with 10 different metastatic solid tumors treated with ICIs targeting programmed-cell death-1 (PD-1)/PD-L1 axis were included in this study. The phenotype of T cells deriving from biopsies obtained prior to treatment initiation and on-treatment was investigaded. Analyses were focused on T cells’ degree of differentiation and activity and how they correlate with transcriptomic changes in the TME.

Results

Data indicate that patients benefitting from ICIs accumulate CD8+central memory T cells. TILs developed an effector-like phenotype over time, which was also associated with a cytolytic gene signature. In terms of modulation of T-cell responses, we observed that high expression of checkpoint molecules pre-treatment (i.e., PD-1, lymphocyte activation gene-3 [LAG-3], B and T-lymphocyte attenuator [BTLA] and T-cell immunoglobulin and mucin domain containing-3 [TIM-3]) was associated with similar gene signature and correlated to treatment benefit. Increasing expression of LAG-3 and BTLA in the CD8 compartment and their co-expression with PD-1 during treatment were, however, a common feature for patients who failed to respond to ICIs.

Conclusions

Besides identifying immune profiles suggestive of response to ICI, our results provide a more nuanced picture regarding expression of checkpoint molecules that goes beyond T-cell anergy.

Section snippets

Background

The presence of immune cells in the tumor microenvironment (TME) has been long known [1], but it is only over the past decades that targeting the immune system as treatment strategy against cancer has gained popularity, especially due to remarkable results yielded by immune checkpoint inhibitors (ICIs) [2], [3], [4].

The rationale behind ICIs is to revert the state of T-cell dysfunctionality where continuous TCR stimulation by chronic antigen exposure in an immune suppressive milieu has led to

Study design

We designed a basket study to investigate shared T-cell phenotypic features and their correlation to genetic changes across different tumor types. The presented study has, therefore, an explorative character and hypothesis-generating intent.

The protocol related to the study was approved by The Danish National Committee on Health Research Ethics (H-16046968) and the Danish Data Protection Agency (RH-2018-44). Written informed consent was obtained prior to any collection and storage of biological

Patients characteristics

From February 2017 to December 2018, 37 patients referred to our service for treatment with ICIs were screened for inclusion in this study. In total, eight patients were excluded due to withdrawal of informed consent (n = 3) and change of therapeutic strategy after first visit (n = 5). The remaining 29 patients were referred for radiologically guided tumor biopsies. The screening process and number of samples used in the study are illustrated in the consort diagram (Supplementary Figure 2). The

Discussion

Despite the remarkable response rates yielded by ICI in cohorts of melanoma [2,27] and non–small cell lung cancer [28], a considerable fraction of patients still do not respond to the therapy [5]. A better understanding of such variable outcomes requires insight into the biological basis for treatment success and failure.

In our study, we explored the functional repertoire of TILs isolated from patients undergoing ICI, prior to treatment start and on-treatment, searching for common elements,

Funding

This study was only possible due to financial support granted by The Danish Cancer Society and Preben & Anna Simonsens Fond.

Disclosure of Competing Interest

The authors have no commercial, proprietary, or financial interest in the products or companies described in this article.

Author Contributions

VL was responsible for conception and design of the study, idea development, inclusion of patients, collection of biological material and clinical data, flow cytometry acquiring/gating, statistical analysis and data visualization, manuscript writing and final approval of the manuscript. AB was responsible for bioinformatic analyses, interpretation of data, critical review and final approval of the manuscript. MH was responsible for assistance with flow cytometry data, critical review and final

References (52)

  • N.A. Rizvi et al.

    Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer

    Science

    (2015)
  • V. Anagnostou et al.

    Evolution of Neoantigen Landscape during Immune Checkpoint Blockade in Non-Small Cell Lung Cancer

    Cancer Discov

    (2017)
  • X. Wang et al.

    PD-L1 expression in human cancers and its association with clinical outcomes

    Onco Targets Ther

    (2016)
  • J.M. Zaretsky et al.

    Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma

    N Engl J Med

    (2016)
  • R.S. Herbst et al.

    Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients

    Nature

    (2014)
  • D.S. Chen et al.

    Elements of cancer immunity and the cancer-immune set point

    Nature

    (2017)
  • N. Riaz et al.

    Tumor and Microenvironment Evolution during Immunotherapy with Nivolumab

    Cell

    (2017)
  • M. Philip et al.

    Chromatin states define tumour-specific T cell dysfunction and reprogramming

    Nature

    (2017)
  • R.D. Schreiber et al.

    Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion

    Science

    (2011)
  • G.P. Dunn et al.

    Cancer immunoediting: from immunosurveillance to tumor escape

    Nat. Immunol

    (2002)
  • A.O. Kamphorst et al.

    Rescue of exhausted CD8 T cells by PD-1-targeted therapies is CD28-dependent

    Science

    (2017)
  • E. Grywalska et al.

    Immune-checkpoint inhibitors for combating T-cell dysfunction in cancer

    Onco Targets Ther

    (2018)
  • D.S. Thommen 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)
  • A. Gros et al.

    PD-1 identifies the patient-specific CD8(+) tumor-reactive repertoire infiltrating human tumors

    J Clin Invest

    (2014)
  • Y. Simoni et al.

    Bystander CD8(+) T cells are abundant and phenotypically distinct in human tumour infiltrates

    Nature

    (2018)
  • Krueger F. Trim Galore [Internet];...
  • Cited by (7)

    View all citing articles on Scopus
    View full text