[HTML][HTML] Interferon-gamma–producing CD8+ tissue resident memory T cells are a targetable hallmark of immune checkpoint inhibitor–colitis

SC Sasson, SM Slevin, VTF Cheung, I Nassiri… - Gastroenterology, 2021 - Elsevier
SC Sasson, SM Slevin, VTF Cheung, I Nassiri, A Olsson-Brown, E Fryer, RC Ferreira
Gastroenterology, 2021Elsevier
Background & Aims The pathogenesis of immune checkpoint inhibitor (ICI)–colitis remains
incompletely understood. We sought to identify key cellular drivers of ICI-colitis and their
similarities to idiopathic ulcerative colitis, and to determine potential novel therapeutic
targets. Methods We used a cross-sectional approach to study patients with ICI-colitis, those
receiving ICI without the development of colitis, idiopathic ulcerative colitis, and healthy
controls. A subset of patients with ICI-colitis were studied longitudinally. We applied a range …
Background & Aims
The pathogenesis of immune checkpoint inhibitor (ICI)–colitis remains incompletely understood. We sought to identify key cellular drivers of ICI-colitis and their similarities to idiopathic ulcerative colitis, and to determine potential novel therapeutic targets.
Methods
We used a cross-sectional approach to study patients with ICI-colitis, those receiving ICI without the development of colitis, idiopathic ulcerative colitis, and healthy controls. A subset of patients with ICI-colitis were studied longitudinally. We applied a range of methods, including multiparameter and spectral flow cytometry, spectral immunofluorescence microscopy, targeted gene panels, and bulk and single-cell RNA sequencing.
Results
We demonstrate CD8+ tissue resident memory T (TRM) cells are the dominant activated T cell subset in ICI-colitis. The pattern of gastrointestinal immunopathology is distinct from ulcerative colitis at both the immune and epithelial-signaling levels. CD8+ TRM cell activation correlates with clinical and endoscopic ICI-colitis severity. Single-cell RNA sequencing analysis confirms activated CD8+ TRM cells express high levels of transcripts for checkpoint inhibitors and interferon-gamma in ICI-colitis. We demonstrate similar findings in both anti–CTLA-4/PD-1 combination therapy and in anti–PD-1 inhibitor-associated colitis. On the basis of our data, we successfully targeted this pathway in a patient with refractory ICI-colitis, using the JAK inhibitor tofacitinib.
Conclusions
Interferon gamma–producing CD8+ TRM cells are a pathological hallmark of ICI-colitis and a novel target for therapy.
Elsevier