Early disappearance of tumor antigen-reactive T cells from peripheral blood correlates with superior clinical outcomes in melanoma under anti-PD-1 therapy

J Immunother Cancer. 2021 Dec;9(12):e003439. doi: 10.1136/jitc-2021-003439.

Abstract

Background: Anti-programmed cell death protein 1 (PD-1) antibodies are now routinely administered for metastatic melanoma and for increasing numbers of other cancers, but still only a fraction of patients respond. Better understanding of the modes of action and predictive biomarkers for clinical outcome is urgently required. Cancer rejection is mostly T cell-mediated. We previously showed that the presence of NY-ESO-1-reactive and/or Melan-A-reactive T cells in the blood correlated with prolonged overall survival (OS) of patients with melanoma with a heterogeneous treatment background. Here, we investigated whether such reactive T cells can also be informative for clinical outcomes in metastatic melanoma under PD-1 immune-checkpoint blockade (ICB).

Methods: Peripheral blood T cell stimulation by NY-ESO-1 and Melan-A overlapping peptide libraries was assessed before and during ICB in two independent cohorts of a total of 111 patients with stage IV melanoma. In certain cases, tumor-infiltrating lymphocytes could also be assessed for such responses. These were characterized using intracellular cytokine staining for interferon gamma (IFN-γ), tumor negrosis factor (TNF) and CD107a. Digital pathology analysis was performed to quantify NY-ESO-1 and Melan-A expression by tumors. Endpoints were OS and progression-free survival (PFS).

Results: The initial presence in the circulation of NY-ESO-1- or Melan-A-reactive T cells which became no longer detectable during ICB correlated with validated, prolonged PFS (HR:0.1; p>0.0001) and OS (HR:0.2; p=0.021). An evaluation of melanoma tissue from selected cases suggested a correlation between tumor-resident NY-ESO-1- and Melan-A-reactive T cells and disease control, supporting the notion of a therapy-associated sequestration of cells from the periphery to the tumor predominantly in those patients benefitting from ICB.

Conclusions: Our findings suggest a PD-1 blockade-dependent infiltration of melanoma-reactive T cells from the periphery into the tumor and imply that this seminally contributes to effective treatment.

Keywords: biomarkers; immunotherapy; lymphocytes; melanoma; programmed cell death 1 receptor; tumor; tumor-infiltrating.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Aged
  • Aged, 80 and over
  • Antigens, Neoplasm / immunology
  • Antigens, Neoplasm / metabolism*
  • Biomarkers, Tumor / immunology
  • Biomarkers, Tumor / metabolism*
  • CD8-Positive T-Lymphocytes / immunology*
  • Female
  • Follow-Up Studies
  • Humans
  • Immune Checkpoint Inhibitors / therapeutic use*
  • Leukocytes, Mononuclear / drug effects
  • Leukocytes, Mononuclear / immunology
  • Leukocytes, Mononuclear / metabolism
  • Leukocytes, Mononuclear / pathology
  • Lymphocytes, Tumor-Infiltrating / immunology
  • MART-1 Antigen / immunology
  • MART-1 Antigen / metabolism*
  • Male
  • Melanoma / drug therapy
  • Melanoma / immunology
  • Melanoma / mortality*
  • Melanoma / pathology
  • Membrane Proteins / immunology
  • Membrane Proteins / metabolism*
  • Middle Aged
  • Prognosis
  • Programmed Cell Death 1 Receptor / antagonists & inhibitors*
  • Survival Rate

Substances

  • Antigens, Neoplasm
  • Biomarkers, Tumor
  • CTAG1B protein, human
  • Immune Checkpoint Inhibitors
  • MART-1 Antigen
  • MLANA protein, human
  • Membrane Proteins
  • PDCD1 protein, human
  • Programmed Cell Death 1 Receptor