Genomic Factors Indicating Sensitivity to IO and Chemotherapy in CCA

March 2024, Vol 5, No 1

There has been an increase in immunotherapy (IO) used for treating advanced cholangiocarcinoma (CCA). However, data are limited to the sensitivity and resistance to chemotherapy and IO in CCA, making it complex to identify predictive biomarkers to guide treatment decision-making.

Roswell Park Comprehensive Cancer Center investigators hypothesized that sensitivity to IO and chemotherapy in programmed death-ligand 1 (PD-L1)–high and PD-L1–low patients can be differentiated using transcriptional signatures.1,2

Additionally, the signatures could facilitate personalized treatment strategies and identify candidates for other novel therapeutic approaches. Riya Jayesh Patel, MD, presented results from this study at ASCO GI.

Transcriptomic signatures related to chemotherapy and IO sensitivity in the CCA cohort of The Cancer Genome Atlas (TCGA-CHOL) were explored. Transcriptional dysregulation was assessed by comparing the transcriptional profiles of the 2 groups.

Differential expression analysis, using limma (a package for analyzing microarray and RNA-sequencing data), was utilized for Gene Set Enrichment Analysis (GSEA) to assess the predicted function of transcriptional dysregulation between the 2 groups. Immune deconvolution was assessed using the Tumor Immune Estimation Resource (TIMER) deconvolution software to evaluate differences in immune infiltration.

The resulting scores from this analysis were used to determine how resistant cell population estimates correlated with immune factors like PD-L1 expression.

Of 36 patients, 33 (91.7%) were of liver and intrahepatic biliary duct origin; 1 (2.8%) was gallbladder primary; and 2 (5.5%) were labeled as “others.”

All patients were divided into 2 groups based on the PD-L1 expression levels. Patients were designated as PD-L1-high (n=11) if their PD-L1 levels had the same or more than average PD-L1 expression and PD-L1-low (n=25) if their PD-L1 levels had the same or more than average expression PD-L1 within the cohort.

Scores from this analysis were used to correlate immune cell estimates with critical immune factors. Highly expressive PD-L1 tumors demonstrated immunosuppressive infiltration.

Using TIMER deconvolution software, several significant correlations were noted between various immune cells: CD4+ and CD8+ T cells (Pearson product-moment correlation coefficient [R]=–0.35; P=.02), CD8+ and neutrophils (R=0.42; P=.009), and CD8+ and myeloid dendritic cells (mDCs; R=0.3348; P=.04).

Additionally, significant correlations were noted between PD-L1 expression and immune cells, including PD-L1 and B cells (R=0.54; P=.0005), macrophages (R=0.41; P=.01), CD8+ T cells (R=0.43; P=.008), neutrophils (R=0.56; P=.0003), and mDCs (R=0.48; P=.002).

GSEA analysis revealed that tumors with high PD-L1 expression exhibit a significant enrichment of unique transcriptional dysregulation patterns.

The genes identified in this study play a crucial role in predicting the effectiveness of chemotherapy drugs used to treat CCA. These genes have also been established as prognostic markers in various other types of cancer.

Additionally, specific metabolic patterns have been observed in tumors with low PD-L1 expression, which is also known to have prognostic value in other cancer types. GSEA also revealed alterations to immune cell pathways associated with PD-L1 expression.

Significant variations were observed in the immune deconvolution scores of CD8+ T cells, macrophages, and neutrophils when comparing PD-L1-high and PD-L1-low patients.

Furthermore, patients with low levels of PD-L1 exhibited notable changes in the genes responsible for metabolic pathways, specifically those involved in regulating cholesterol, amino acid transportation, and cellular respiration.

The PD-L1-high group showed enrichment of immune signatures, and the PD-L1-low group showed enrichment of immune signatures in the TCGA-CHOL patients.

Patients with CCA with high levels of PD-L1 displayed expected differential enrichment of immune signatures. It is reported for the first time that CCA PD-L1-low patients are uniquely enriched for differential metabolic signatures.

If validated in cohorts treated with IO, these findings could offer valuable insights and serve as evidence supporting the effectiveness of metabolism-targeted therapies in overcoming therapeutic resistance in CCA and potentially facilitating personalized treatment strategies for other novel therapeutic approaches.2

References

  1. Kawamura E, Matsubara T, Kawada N. New era of immune-based therapy in intrahepatic cholangiocarcinoma. Cancers (Basel). 2023;15(15):3993.
  2. Patel RJ, Rosario S, Sonti S, et al. Genomic predictors of sensitivity to chemotherapy and immunotherapy in cholangiocarcinoma. San Francisco, CA, & online: presented at ASCO Gastrointestinal Cancers Symposium; January 18-20, 2024: poster F7.

Related Items

Tasurgratinib for Patients With FGFR2 Gene Fusion–Positive CCA: A Phase 2 Study
March 2024, Vol 5, No 1
Junji Furuse, MD, PhD, presented the results of a phase 2 study of tasurgratinib efficacy on patients with FGFR2 fusion-positive cholangiocarcinoma following gene fusion status confirmation by fluorescence in situ hybridization.
Safety and Efficacy of Telotristat Ethyl Plus First-Line Chemotherapy in Patients With Advanced BTC: A Phase 2, Open-Label Study
March 2024, Vol 5, No 1
Richard Kim, MD, presented the results of a phase 2, open-label study of patients who underwent telotristat ethyl plus first-line chemotherapy for the treatment of advanced biliary tract cancer.
Efficacy and Safety of Brigimadlin (BI 907828) in Patients With Advanced BTC: Data From 2 Phase 1a/1b Dose-Escalation/Expansion Trials
March 2024, Vol 5, No 1
Teresa Macarulla, MD, PhD, presented the results of 2 phase 1a/1b dose-escalation/expansion trials, studies that measured the efficacy and safety of brigimadlin in patients with advanced biliary tract cancer.
COMPANION-002: A Phase 2/3 Randomized Study Design of CTX-009 Combination in Second-Line BTC
March 2024, Vol 5, No 1
A recombinant bispecific antibody, CTX-009, is discussed regarding its role in an ongoing phase 2/3 open-label, randomized, controlled study—a study being conducted to measure the efficacy of CTX-009 in previously treated, advanced, or metastatic biliary tract cancer.
A Phase 2 Clinical Trial of Anlotinib Plus TQB2450 (PD-L1 Blockade) Plus Nab-Paclitaxel and Cisplatin as First-Line Treatment for Advanced BTC
March 2024, Vol 5, No 1
Cholangiocarcinoma experts presented the preliminary results of a phase 2 clinical trial regarding anlotinib plus TQB2450, nab-paclitaxel, and cisplatin—a study conducted for the treatment of advanced biliary tract cancer.
Preliminary Results of a Real-World Study of the Safety and Efficacy of Surufatinib in BTC
March 2024, Vol 5, No 1
Zongli Zhang, MD, PhD, presented the preliminary results of an ongoing single-arm, multicenter, open-label, real-world study analyzing the efficacy and safety of surufatinib as a second-line treatment option for patients with biliary tract cancer.
Examining Real-World Testing, Treatment Patterns, and Outcomes After Liquid Biopsy in aCCA
March 2024, Vol 5, No 1
Amit Mahipal, MD, presented results from a real-world data study examining the rates of molecular alterations detected using circulating tumor DNA (ctDNA) for patients receiving ivosidenib following circulating tumor ctDNA-detected IDH1 mutations.
A Phase 2 Primary Analysis of Tislelizumab Plus Lenvatinib and GEMOX as Conversion Therapy in Potentially Resectable Locally Advanced BTC (ZSAB-TransGOLP)
March 2024, Vol 5, No 1
Jia Fan, PhD, presented results from a phase 2 trial that investigated the efficacy and safety of lenvatinib and a programmed cell death protein-1 antibody as conversion therapy for the treatment of potentially resectable and locally advanced biliary tract cancer.
Role of CD27 Agonist in Combination With PD-L1 and MEK Inhibition on Antitumor Effect and CD8+ T Cells: Expanding Immunotherapy Options in CCA
March 2024, Vol 5, No 1
Frances J. Bennett, MD, presented the results of a phase 2 trial that tested the antitumor effect of dual programmed death-ligand 1 plus mitogen-activated protein kinase inhibition in patients with advanced biliary tract cancer.
Phase 3 Study of Tinengotinib Versus Physician’s Choice in Patients With FGFR-Altered, Chemotherapy- and FGFR Inhibitor-R/R CCA: Study Design of FIRST-308
March 2024, Vol 5, No 1
FIRST-308 is an open-label, phase 2, randomized, global, multicenter study that will evaluate the efficacy and safety of oral tinengotinib versus physician’s choice in patients with fibroblast growth factor receptor (FGFR)-altered, chemotherapy- and FGFR inhibitor-refractory/relapsed cholangiocarcinoma—this article will review the study’s eligibility criteria, primary endpoints, and more.

Subscribe Today!

To sign up for our newsletter or print publications, please enter your contact information below.

I'd like to receive: