Prevalence of established and emerging biomarkers of immune checkpoint inhibitor response in advanced hepatocellular carcinoma

The clinical deployment of immune checkpoint inhibitors (ICIs) has created a tandem drive for the identification of biomarkers linked to benefit. Comprehensive genomic profiling was performed to evaluate the frequency of genomic biomarkers of ICI response in 755 patients with advanced hepatocellular carcinoma (HCC). Median age was 62 years’ old, 73% were male, 46% had extrahepatic disease, 107 had documented hepatitis C, 96 had hepatitis B and 4 patients were coinfected. Median tumor mutation burden (TMB) was 4 mutations/Mb and only 6 tumors (0.8%) were TMB-high. Out of 542 cases assessed for microsatellite instability (MSI), one (0.2%) was MSI-high and TMB-high. Twenty-seven (4%) patients had POLE/D alterations. One patient had a pathogenic POLE R762W mutation but TMB was 4 mutations/Mb. Forty percent had DNA damage response gene alterations. In a small case series (N=17) exploring the relationship between biomarkers and ICI response, one patient (TMB 15 mutations/Mb, MSI-low) had a sustained complete response to nivolumab lasting > 2 years. Otherwise there were no significant genomic or TMB differences between responders, progressors, and those with stable disease. Overall, markers of genomic instability were infrequent in this cohort. Larger clinically annotated datasets are needed to explore genomic and non-genomic determinants of ICI response in HCC.

[1]  T A Chan,et al.  Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic , 2018, Annals of oncology : official journal of the European Society for Medical Oncology.

[2]  M. Kudo,et al.  Pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib (KEYNOTE-224): a non-randomised, open-label phase 2 trial. , 2018, The Lancet. Oncology.

[3]  M. Berger,et al.  Alterations in DNA Damage Response and Repair Genes as Potential Marker of Clinical Benefit From PD-1/PD-L1 Blockade in Advanced Urothelial Cancers. , 2018, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[4]  Laurence Zitvogel,et al.  Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors , 2018, Science.

[5]  E. Jaffee,et al.  Tumor Mutational Burden and Response Rate to PD-1 Inhibition. , 2017, The New England journal of medicine.

[6]  N. Chaput,et al.  Hypermutated tumours in the era of immunotherapy: The paradigm of personalised medicine. , 2017, European journal of cancer.

[7]  C. Desbois-Mouthon,et al.  Low Levels of Microsatellite Instability at Simple Repeated Sequences Commonly Occur in Human Hepatocellular Carcinoma. , 2017, Cancer genomics & proteomics.

[8]  P. Stephens,et al.  Tumor Mutational Burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers , 2017, Molecular Cancer Therapeutics.

[9]  M. Kudo,et al.  Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial , 2017, The Lancet.

[10]  Levi Garraway,et al.  Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden , 2017, Genome Medicine.

[11]  Jun Gong,et al.  Response to PD-1 Blockade in Microsatellite Stable Metastatic Colorectal Cancer Harboring a POLE Mutation. , 2017, Journal of the National Comprehensive Cancer Network : JNCCN.

[12]  A. Villanueva,et al.  Genetic profiling of hepatocellular carcinoma using next-generation sequencing. , 2016, Journal of hepatology.

[13]  M. Dolled-Filhart,et al.  Development of a Companion Diagnostic PD-L1 Immunohistochemistry Assay for Pembrolizumab Therapy in Non–Small-cell Lung Cancer , 2016, Applied immunohistochemistry & molecular morphology : AIMM.

[14]  J. Taube,et al.  Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy , 2016, Nature Reviews Cancer.

[15]  M. Postow,et al.  Emerging Tissue and Blood-Based Biomarkers that may Predict Response to Immune Checkpoint Inhibition , 2016, Current Oncology Reports.

[16]  Ian Tomlinson,et al.  A panoply of errors: polymerase proofreading domain mutations in cancer , 2016, Nature Reviews Cancer.

[17]  S. Millis,et al.  Comprehensive multiplatform biomarker analysis of 350 hepatocellular carcinomas identifies potential novel therapeutic options , 2016, Journal of surgical oncology.

[18]  J. Prieto,et al.  Immunological landscape and immunotherapy of hepatocellular carcinoma , 2015, Nature Reviews Gastroenterology &Hepatology.

[19]  Jorge Sabbaga,et al.  Comprehensive cancer-gene panels can be used to estimate mutational load and predict clinical benefit to PD-1 blockade in clinical practice , 2015, Oncotarget.

[20]  J. Zucman‐Rossi,et al.  Genetic Landscape and Biomarkers of Hepatocellular Carcinoma. , 2015, Gastroenterology.

[21]  B. Vogelstein,et al.  PD-1 blockade in tumors with mismatch repair deficiency. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[22]  J. Wolchok,et al.  Genetic basis for clinical response to CTLA-4 blockade in melanoma. , 2014, The New England journal of medicine.

[23]  J. Taube,et al.  Association of PD-1, PD-1 Ligands, and Other Features of the Tumor Immune Microenvironment with Response to Anti–PD-1 Therapy , 2014, Clinical Cancer Research.

[24]  Peter Donnelly,et al.  Germline mutations affecting the proofreading domains of POLE and POLD1 predispose to colorectal adenomas and carcinomas , 2013, Nature Genetics.

[25]  David C. Smith,et al.  Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. , 2012, The New England journal of medicine.

[26]  N. Yoo,et al.  Somatic mutations of the KEAP1 gene in common solid cancers , 2012, Histopathology.