Despite advances in diagnostic imaging, surgical techniques, and systemic therapy, gastric cancer (GC) is the third leading cause of cancer-related death worldwide. Unfortunately, molecular heterogeneity and, consequently, acquired resistance in GC are the major causes of failure in the development of biomarker-guided targeted therapies. However, by showing promising survival benefits in some studies, the recent emergence of immunotherapy in GC has had a significant impact on treatment-selectable procedures. Immune checkpoint inhibitors (ICIs), widely indicated in the treatment of several malignancies, target inhibitory receptors on T lymphocytes, including the programmed cell death protein (PD-1)/programmed death-ligand 1 (PD-L1) axis and cytotoxic T-lymphocyte-associated protein 4 (CTLA4), and release effector T-cells from negative feedback signals.
1. Introduction
Despite advances in diagnostic imaging, surgical techniques, and systemic therapy, gastric cancer (GC) is one of the most common cancers, with a high mortality rate from cancer around the world
[1][2]. Unfortunately, morphological and molecular heterogeneity and, consequently, acquired resistance in GC are the major causes of failure in the development of biomarker-guided targeted therapies
[2][3]. However, by showing promising survival benefits in some studies, the recent emergence of immunotherapy in GC has had a significant impact on treatment-selectable procedures (
Figure 1)
[4][5][6][7][8]. To protect the host from external antigens and autoimmune reactions, the immune system is regulated via a number of receptor–ligand interactions
[2][9][10]. Immune checkpoint inhibitors (ICIs), widely indicated in the treatment of several malignancies, target inhibitory receptors on T lymphocytes, including the programmed cell death protein (PD-1)/programmed death-ligand 1 (PD-L1) axis and cytotoxic T-lymphocyte-associated protein 4 (CTLA4), and release effector T-cells from negative feedback signals
[2][10][11]. Currently, with limited samples, it is important to identify target biomarkers for predicting response to ICIs
[12][13]. PD-L1 immunohistochemistry (IHC), microsatellite instability (MSI)/mismatch repair (MMR), Epstein–Barr virus (EBV), and tumor mutational burden (TMB) have been proposed as predictive biomarkers to predict response to ICIs in patients with GC
[12][14].
Figure 1. Timeline of major milestones for immunotherapy and U.S. Food and Drug Administration (FDA)-approved Immune checkpoint inhibitors in gastric cancers. MSI-H, microsatellite instability-high; MMR-D, mismatch repair deficiency; GEJ, gastroesophageal junction; PD-L1, programmed death ligand 1; CPS, combined positive score; TMB-H, tumor mutational burden-high; mut/Mb, mutations/megabase; GC, gastric cancer; GEJC, gastroesophageal junction cancer; HER-2, human epidermal growth factor receptor 2.
2. Rationale and Performance
Honjo’s group first discovered PD-1 in 1992
[11][15]. PD-1 is mainly expressed in activated cytotoxic T-cells and other immune cells
[5][11][16]. It is a cell surface (or transmembrane) protein encoded by the CD274 gene
[2][17]. The interaction of PD-1, expressed on cytotoxic T lymphocytes, with PD-L1 on antigen-presenting cells is one of the main mechanisms of immune modulation, thereby allowing T-cell inactivation and tumor immune evasion
[2][10][16][18]. Tumor cells with mutable neoantigens are recognized as ‘non-self’ by the immune system and are selectively targeted and removed by inducing an immune response
[10]. To avoid removal, tumor cells can upregulate PD-L1 and PD-L2 expression following exposure to interferon-γ and other signaling and cytokines
[10][11][16][17][19]. Furthermore, PD-L1 expression is increased in some immune cells within the tumor microenvironment, including dendritic cells, macrophages, antigen-presenting cells, and T-cells
[17][20]. By this principle, the blockade of PD-1/PD-L1 interaction by monoclonal antibodies against either PD-1 (pembrolizumab and nivolumab) or PD-L1 (durvalumab, atezolizumab, and avelumab) appears to be a logical therapeutic strategy, particularly for a highly antigenic solid tumor, including GC
[10][21].
The phase 2 KEYNOTE-059 trial (NCT#02335411), which enrolled 259 patients with locally advanced or metastatic gastric or gastroesophageal junction (G/GEJ) adenocarcinoma suggested the safety and effectiveness of pembrolizumab as a third-line treatment
[4][11]. In this single-arm, multicohort trial, the objective response rate (ORR) was 11.6% (30 of 259 patients), and complete responses (CRs) were noted in 2.3% of patients (6 of 259); the median (range) response duration was 8.4 (1.6+–17.3+) months
[4][11]. Among 259 patients, 148 (57.1%) with PD-L1-positive tumors (combined positive score [CPS] ≥1, evaluated using the PD-L1 22C3 pharmDx Kit) had ORRs and CRs of 15.5% (23 of 148) and 2.0% (3 of 148), respectively. The median response duration was 16.3 (1.6+–17.3+) months. Based on these results, pembrolizumab, in 2017, was granted accelerated Food and Drug Administration (FDA)-approval as a third-line therapy for patients with locally advanced or metastatic G/GEJ adenocarcinoma whose tumors express a CPS of ≥1 using the PD-L1 immunohistochemistry (IHC) 22C3 pharmDx assay
[2][18]. Notably, pembrolizumab was approved, along with a companion diagnostic test, the PD-L1 IHC 22C3 pharmDx assay, for use on the Dako Autostainer Link 48 platform
[4][18]. However, after accelerating FDA approval, pembrolizumab failed to demonstrate significant survival improvements in the following phase 3 trials (KEYNOTE-061 [NCT#02370498; efficacy as a second-line therapy] and KEYNOTE-062 [NCT#02494583; efficacy as a first-line therapy]) in patients with advanced G/GEJ adenocarcinoma showing a PD-L1 CPS of ≥1
[5][6][11][18].
Another phase 3 CheckMate-649 trial (NCT#02872116), which enrolled 1581 patients with untreated, unresectable, Human Epidermal Growth Factor Receptor 2 (HER2)-negative G/GEJ, or esophageal adenocarcinoma, demonstrated the acceptable safety profile and efficacy of nivolumab as a first-line treatment
[7][18]. The CheckMate-649 trial demonstrated significant improvements in the overall survival (OS) and progression-free survival (PFS) for patients with a CPS of ≥5 (955 of 1581; 60.4%) determined using the PD-L1 28-8 pharmDx kit on the Autostainer Link 48 platform
[7]. Specifically, the median OS was 14.4 months (95% confidence interval [CI]: 13.1–16.2) in the nivolumab plus chemotherapy group (n = 789) versus 11.1 months (95% CI: 10.0–12.1) in the chemotherapy-alone group (n = 792) (
p < 0.0001)
[7]. Moreover, the median PFS was 7.7 months (95% CI: 7.0–9.2) in the nivolumab plus chemotherapy arm versus 6.0 months (95% CI: 5.6–6.9) in the chemotherapy alone arm (
p < 0.0001)
[7]. Based on these results, in 2021, the FDA-approved, nivolumab plus fluoropyrimidine- and platinum-based chemotherapy as a first-line therapy for HER2-negative advanced or metastatic G/GEJ and esophageal adenocarcinoma
[2]. Nivolumab was approved along with a companion diagnostic test, the PD-L1 IHC 28-8 pharmDx assay, for use on the Dako Autostainer Link 48 platform
[2][18]. However, the phase 3 ATTRACTION-4 trial demonstrated that first-line nivolumab plus oxaliplatin-based chemotherapy resulted in a significant improvement in PFS, but not OS, in Asian patients with untreated, HER2-negative, unresectable advanced or recurrent G/GEJ adenocarcinoma with a tumor proportion score (TPS) of ≥1 (114 of 724; 15.7%)
[22]. In this trial, PD-L1 expression was evaluated using the PD-L1 IHC 28-8 pharmDx assay and was scored using TPS, not CPS
[22].
Recently, the interim results of the phase 3 KEYNOTE-811 trial (NCT#03615326) in enrolled patients with unresectable or metastatic, HER2-positive G/GEJ adenocarcinoma showed that first-line pembrolizumab plus trastuzumab and chemotherapy significantly reduced tumor size, induced CRs in some participants, and significantly improved ORR
[8]. In this trial, PD-L1 expression was evaluated using the PD-L1 IHC 22C3 pharmDx assay and was scored using CPS (patients with a CPS of ≥1: 582 of 692, 84.1%)
[8]. Although KEYNOTE-811 trial has proceeded, pembrolizumab plus trastuzumab and chemotherapy could potentially be a new first-line treatment option for patients with unresectable advanced or metastatic, HER2-positive G/GEJ adenocarcinoma.
This entry is adapted from the peer-reviewed paper 10.3390/diagnostics13172782