Large-scale transcriptomic profiling of the tumor immune microenvironment in ALK+ lung cancer.

Authors

null

Vincent K. Lam

The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD

Vincent K. Lam , Archana Balan , Qingfeng Zhu , Joseph Christopher Murray , Kristen A. Marrone , Susan Combs Scott , Josephine Louella Feliciano , Christine L. Hann , David S. Ettinger , Kellie Nicole Smith , Patrick M. Forde , Julie R. Brahmer , Benjamin Philip Levy , Andrew Elliott , Ari VanderWalde , Matthew James Oberley , Stephen V. Liu , Patrick C. Ma , Robert A. Anders , Valsamo Anagnostou

Organizations

The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, The Sidney Kimmel Comprehensive Cancer Center and Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, MD, Johns Hopkins Kimmel Cancer Center, Baltimore, MD, Johns Hopkins University School of Medicine, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, Johns Hopkins Kimmel Cancer Center and Bloomberg-Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, Caris Life Sciences, Phoenix, AZ, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, Penn State Milton S. Hershey Medical Center, Hershey, PA, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD

Research Funding

Other Foundation
Lung Cancer Foundation of America and International Lung Cancer Foundation

Background: Patients (pts) with ALK+ Non-Small Cell Lung Cancer (NSCLC) do not derive significant clinical benefit from immune checkpoint inhibitors. To better understand this lack of immunotherapy sensitivity, we aimed to characterize major immune components of the tumor microenvironment (TME) by comprehensive transcriptomic and immunohistochemistry (IHC) analyses. Methods: We analyzed NGS data from 5490 NSCLC pts that underwent DNA (592 Gene Panel, NextSeq, or WES, NovaSeq) and RNA (NovaSeq, WTS) sequencing at Caris Life Sciences (Phoenix, AZ). 374 ALK-rearranged cases were evaluated, along with 3169 KRAS-mut (STK11/KEAP1-wt) and 1947 EGFR-mut cases serving as comparators with known heterogenous and inert immune TMEs, respectively. PD-L1 (22C3) was evaluated by IHC. Immune cell fractions were inferred using quanTIseq. Gene expression profiles were analyzed for a T cell-inflamed signature (TIS) predictive of response to immunotherapy and for other immune modulatory genes such as IFNG, GZMB, TGFB1, and those of the adenosine pathway (CD73/NT5E, CD39/ENTPD1, ADORA1, ADORA2A/B). A significant difference between genomic subgroups was defined as fold-change > 1.2. In an independent cohort of 14 ALK+ NSCLC pts, density and spatial organization of CD4+ and CD8+ T cells, Tregs, major myeloid lineage cells, PDL1, and CD73 were assessed by quantitative IHC. Results:ALK+ tumors were associated with high PD-L1 (≥50%) expression (40% vs 47% for KRAS-mut vs 18% for EGFR-mut, p < 0.001) and low TMB (median 3 mut/MB vs 9 for KRAS-mut vs 4 for EGFR-mut, p < 0.001). The abundance of CD8+ T cells (fold-change -1.3, p < 0.001), Tregs (fold-change -1.2, p < 0.001), M2 macrophages (fold-change 1.2, p < 0.001), and CD4+ T cells (fold-change 1.9, p < 0.001) differed from KRAS-mut; notably, similar to EGFR-mut. In ALK+ tumors, IFNG (fold-change -1.5, p < 0.001), GZMB (fold-change -1.6, p < 0.001), TGFB (fold-change -1.3, p < 0.001), LAG-3 (fold-change -1.4 p < 0.001), CD73/NT53 (fold-change -1.7 p < 0.001), and ADORA2A (fold-change -1.4, p < 0.001) were decreased while ADORA1 (fold-change 1.3, p < 0.001) was increased compared to KRAS-mut. EML4-ALK comprised 94.7% of the ALK+ tumors and distribution of EML4-ALK variants was consistent with prior reports (e.g. v1 35.6%, v3 35.1%). Immune cell fractions and immune-related gene expression did not vary significantly between major variant subgroups (v1 vs non-v1, and v3 vs non-v3, p > 0.05). Conclusions: To our knowledge this is the largest transcriptomic analysis of the ALK+ NSCLC TME. Despite high levels of PD-L1, ALK+ tumors exhibit multiple features of an inert immune TME, primarily characterized by low TMB and decreased CD8+ T cells and immune activation markers. Our findings suggest that, while immunosuppressive factors such as M2 macrophages and adenosine signaling may also be targeted, strategies to enhance immunogenicity are critical for an effective immune response in ALK+ NSCLC.

Disclaimer

This material on this page is ©2024 American Society of Clinical Oncology, all rights reserved. Licensing available upon request. For more information, please contact licensing@asco.org

Abstract Details

Meeting

2023 ASCO Annual Meeting

Session Type

Poster Discussion Session

Session Title

Lung Cancer—Non-Small Cell Metastatic

Track

Lung Cancer

Sub Track

Biologic Correlates

Citation

J Clin Oncol 41, 2023 (suppl 16; abstr 9020)

DOI

10.1200/JCO.2023.41.16_suppl.9020

Abstract #

9020

Poster Bd #

8

Abstract Disclosures

Similar Abstracts

First Author: Stephanie Leigh Alden

First Author: Vincent Lacasse

First Author: Vincent Lacasse

First Author: Sarabjot Pabla