Immunotherapy in Cholangiocarcinoma: Coming of Age

June/July 2022, Vol 3, No 2
Richard Kim, MD
Service Chief, Medical Gastrointestinal Oncology
Senior Member, Gastrointestinal Oncology Department
Moffitt Cancer Center
Professor, Oncology
University of South Florida College of Medicine
Tampa, FL
Amit Mahipal, MBBS, MPH
Department of Oncology, Mayo Clinic
Rochester, MN

Introduction

Cholangiocarcinoma (CCA) is a relatively rare, aggressive malignancy originating from epithelial cells of the biliary duct that constitutes approximately 3% of the gastrointestinal malignancies. CCAs are anatomically classified into intrahepatic, perihilar, and extrahepatic subtypes, each with a distinct epidemiology and molecular pathogenesis.1 There has been increasing incidence of cholangiocarcinoma globally, including in the United States. A majority of patients present at an advanced stage requiring systemic therapy. Recent advances in genomic profiling and critical understanding of pathophysiology have led to the development of multiple targeted therapies in addition to chemotherapeutic agents.2

Immunotherapy has dramatically altered the treatment paradigm of multiple cancers. In CCA, the role of immunotherapy remains undefined. Immunotherapy is considered a potential target as chronic inflammation including primary sclerosing cholangitis seems to play a role in the development of CCA. CCAs are characterized by a desmoplastic tumor environment consisting of stromal cancer-associated fibroblasts and immune cells, including tumor-associated macrophages, natural killer cells, and T-cells.3 PD-L1 expression has been reported in cholangiocarcinoma neoplastic cells with a wide range of 9% to 72%, as well as inflammatory cell aggregates.4-6 Deficiency in mismatch repair proteins has been reported in a minority of CCA patients suggesting a potential role for immunotherapy. In general, patients with CCA have low tumor burden but there is a subset of patients who have higher tumor mutational burden (TMB) which might be more responsive to immunotherapy.7

Immune checkpoint inhibitors

Single-agent immune checkpoint inhibitors (ICIs) have demonstrated limited activity with an overall response rate (ORR) of 5% to 25% in unselected populations with refractory CCA (Table 1). In the KEYNOTE-028 basket trial, 24 patients with PDL-1–positive biliary tract cancers (BTCs) were treated with pembrolizumab and an ORR of 13% was reported.8 In the larger KEYNOTE-158 trial, 104 patients with advanced, refractory BTCs were enrolled. ORR was only 5.8% with median progression-free survival (PFS) and overall survival (OS) of 2 months and 7.4 months, respectively. Patients with PD-L1–positive tumors had a numerically higher ORR (6.6% vs 2.9%). In a phase 2 trial of 54 patients with advanced BTC, treatment with nivolumab resulted in an ORR of 22%.9 Bintrafusp alfa, a bifunctional fusion protein targeting TGF-beta and PD-L1 demonstrated similar activity, with an ORR of 22% and median PFS of 3.7 months.10

Table 1

Dual checkpoint inhibitor therapy has also been evaluated in CCA. The combination of durvalumab plus tremelimumab demonstrated an ORR of 10.8%, which was higher than that seen with durvalumab alone (4.8%). The combination of nivolumab plus ipilimumab yielded response rates of 23% with a median PFS of 2.9 months. Unfortunately, with the exception of MSI-high tumors, treatment with ICI rarely leads to long-term disease control in patients with CCA.

Combination of ICI and chemotherapy

ICI plus chemotherapy combination therapy has been proved to be safe and feasible in patients with advanced CCA (Table 2). Nivolumab in combination with gemcitabine plus cisplatin as first-line therapy resulted in median PFS and OS of 8.8 and 10.6 months, respectively.11 A phase 2 trial evaluated the combination of durvalumab with and without tremelimumab and gemcitabine plus cisplatin in the first-line setting.12 Patients receiving durvalumab plus chemotherapy achieved a 100% disease control rate with a median PFS of 11 months. The addition of tremelimumab did not confer any benefit. Bintrafusp alfa was evaluated in combination with gemcitabine plus cisplatin in a randomized phase 2/3 trial. Unfortunately, the trial did not meet its primary end point of OS.

Table 2

Prior immunotherapy plus chemotherapy studies led to a large phase 3 randomized trial, TOPAZ-1 (N = 685), evaluating the role of the addition of durvalumab to gemcitabine plus cisplatin.13 Standard-of-care chemotherapy was continued for up to 8 cycles in both arms and patients received durvalumab or placebo as maintenance therapy. The study met its primary end point with a median OS of 12.8 months in durvalumab plus chemotherapy compared with 11.5 months in the chemotherapy alone arm (hazard ratio, 0.80; 95% confidence interval, 0.66-0.97; P = .021). Moreover, both the 12-month OS rates (54.1% vs 48%) and the 24-month OS rates (24.9% vs 10%) were higher in the immunotherapy plus chemotherapy arm. Median PFS was also longer in the combination arm compared with the chemotherapy arm (7.2 vs 5.7 months; P = .75). The benefit of the addition of durvalumab was observed in all subgroups but was more pronounced in patients of Asian descent and having locally advanced cancer rather than metastatic disease. ORR was 26.7% in the durvalumab-chemotherapy arm and 18.7% in the chemotherapy-only arm. PD-L1 expression at various cutoffs was not predictive of efficacy outcomes. There was no significant increase in frequency or severity of adverse events with the addition of durvalumab. At the time of presentation, data were not reported for specific subsets, including viral hepatitis, liver fluke disease, and nonalcoholic steatohepatitis. Furthermore, the OS curves between the 2 arms appear to begin separating at 6 months at the same time chemotherapy was discontinued from both arms.

Other immunotherapeutic agents

ICI is being evaluated in combination with other therapies, including PARP inhibitors, anti-angiogenic agents, and tyrosine kinase inhibitors. Cancer vaccines utilizing tumor-specific antigens based on peptides and dendritic cells to prime T-cells are also being evaluated. Wilms tumor-1 and Mucin-1 are both overexpressed in CCA and are used as targets for vaccine therapy. Similarly, adoptive cell therapy using chimeric antigen receptors and T-cell receptors are also under investigation for patients with CCA.

Conclusion

The role of immunotherapy in CCA continues to evolve. The TOPAZ-1 trial has established the role of durvalumab in addition to chemotherapy as first-line therapy for patients with advanced CCA. The survival curves in the trial separated after cessation of chemotherapy, which may suggest a greater role for immunotherapy as maintenance therapy. There is an urgent need to develop biomarkers that can help in selecting patients to maximize benefit. Future novel immunotherapeutic approaches including cancer vaccines and CAR-T therapies could potentially revolutionize this treatment approach for CCA, which was traditionally not considered as particularly responsive to immunotherapeutic agents.

References

  1. Valle JW, Kelley RK, Nervi B, et al. Biliary tract cancer. Lancet. 2021;397:428-444.
  2. Tella SH, Kommalapati A, Borad MJ, Mahipal A. Second-line therapies in advanced biliary tract cancers. Lancet Oncol. 2020;21:e29-e41.
  3. Kang S, El-Rayes BF, Akce M. Evolving role of immunotherapy in advanced biliary tract cancers. Cancers (Basel). 2022;14.
  4. Ahn S, Lee JC, Shin DW, et al. High PD-L1 expression is associated with therapeutic response to pembrolizumab in patients with advanced biliary tract cancer. Sci Rep. 2020;10:12348.
  5. Gani F, Nagarajan N, Kim Y, et al. Program death 1 immune checkpoint and tumor microenvironment: implications for patients with intrahepatic cholangiocarcinoma. Ann Surg Oncol. 2016;23:2610-2617.
  6. Sabbatino F, Villani V, Yearley JH, et al. PD-L1 and HLA class I antigen expression and clinical course of the disease in intrahepatic cholangiocarcinoma. Clin Cancer Res. 2016;22:470-478.
  7. Jusakul A, Cutcutache I, Yong CH, et al. Whole-genome and epigenomic landscapes of etiologically distinct subtypes of cholangiocarcinoma. Cancer Discov. 2017;7:1116-1135.
  8. Piha-Paul SA, Oh DY, Ueno M, et al. Efficacy and safety of pembrolizumab for the treatment of advanced biliary cancer: results from the KEYNOTE-158 and KEYNOTE-028 studies. Int J Cancer. 2020;147:2190-2198.
  9. Kim RD, Chung V, Alese OB, et al. A phase 2 multi-institutional study of nivolumab for patients with advanced refractory biliary tract cancer. JAMA Oncol. 2020;6:888-894.
  10. Yoo C, Oh DY, Choi HJ, et al. Phase I study of bintrafusp alfa, a bifunctional fusion protein targeting TGF-beta and PD-L1, in patients with pretreated biliary tract cancer. J Immunother Cancer. 2020;8.
  11. Ueno M, Ikeda M, Morizane C, et al. Nivolumab alone or in combination with cisplatin plus gemcitabine in Japanese patients with unresectable or recurrent biliary tract cancer: a non-randomised, multicentre, open-label, phase 1 study. Lancet Gastroenterol Hepatol. 2019;4:611-621.
  12. Oh D-Y, Lee K-H, Lee D-W, et al. Phase II study assessing tolerability, efficacy, and biomarkers for durvalumab (D) ± tremelimumab (T) and gemcitabine/cisplatin (GemCis) in chemo-naïve advanced biliary tract cancer (aBTC). J Clin Oncol. 2020;38:4520-4520.
  13. Oh D-Y, He AR, Qin S, et al. A phase 3 randomized, double-blind, placebo-controlled study of durvalumab in combination with gemcitabine plus cisplatin (GemCis) in patients (pts) with advanced biliary tract cancer (BTC): TOPAZ-1. J Clin Oncol. 2022;40:378-378.
  14. Yarchoan M, Cope L, Ruggieri AN, et al. Multicenter randomized phase II trial of atezolizumab with or without cobimetinib in biliary tract cancers. J Clin Invest. 2021;131.
  15. Ioka T, Ueno M, Oh D-Y, et al. Evaluation of safety and tolerability of durvalumab (D) with or without tremelimumab (T) in patients (pts) with biliary tract cancer (BTC). J Clin Oncol. 2019;37:387-387.
  16. Villanueva L, Lwin Z, Chung HC, et al. Lenvatinib plus pembrolizumab for patients with previously treated biliary tract cancers in the multicohort phase II LEAP-005 study. J Clin Oncol. 2021;39:321.
  17. Chen X, Wu X, Wu H, et al. Camrelizumab plus gemcitabine and oxaliplatin (GEMOX) in patients with advanced biliary tract cancer: a single-arm, open-label, phase II trial. J Immunother Cancer. 2020;8.
  18. Li W, Yu Y, Xu X, et al. Toripalimab with chemotherapy as first-line treatment for advanced biliary tract tumors: Update analytic results of an open-label phase II clinical study (JS001-ZS-BC001). J Clin Oncol. 2021;39:e16170-e.
  19. Feng K, Liu Y, Zhao Y, et al. Efficacy and biomarker analysis of nivolumab plus gemcitabine and cisplatin in patients with unresectable or metastatic biliary tract cancers: results from a phase II study. J Immunother Cancer. 2020;8.

Related Items

Combined Hepatocellular Cholangiocarcinoma
By Jennifer J Knox, MD, MSc, FRCPC; Gordon T Moffat, MD, FRCPC
June 2023, Vol 4, No 2
Combined hepatocellular cholangiocarcinoma (cHCC-CCA) is a unique malignancy comprised of both hepatocellular carcinoma (HCC) from hepatocytes and cholangiocarcinoma (CCA) from bile duct epithelial cells within the same tumor. Initially thought to be a rare entity, the incidence is increasing (up to 14%), yet recognition remains low.
2022 Year in Review: Cholangiocarcinoma
By Richard Kim, MD
2022 Year in Review: Cholangiocarcinoma
In 2022, the COVID-19 pandemic continued to impact the practice of medicine and dissemination of treatment advances presented in scientific forums. Adapting to the changes, societies such as the American Society of Clinical Oncology, Gastrointestinal Cancers Symposium, and European Society for Medical Oncology have adopted virtual and hybrid formats that deliver cutting-edge research in the advancement of oncology care. Recognizing the challenges of the virtual and hybrid formats in terms of reach and impact, we are bringing the Year in Review series that seeks to disseminate key information on treatment advances to clinicians in a timely and effective manner.
Surgical Management and Neoadjuvant Therapy for Cholangiocarcinoma
By Flavio G. Rocha, MD, FACS, FSSO
September 2022, Vol 3, No 3
Surgery remains the only potentially curative modality for cholangiocarcinoma (CCA) in any site of origin.
Key Abstracts Presented at the European Society for Medical Oncology (ESMO) 2021 Annual Meeting
By Richard Kim, MD
Videos
Richard Kim, MD, from the Moffitt Cancer Center, University of South Florida College of Medicine, provides expert commentary on key abstracts and posters presented at the 2021 annual meeting of the European Society for Medical Oncology. Dr Kim will highlight new data from 11 presentations on interventional modalities for patients with cholangiocarcinoma.
Managing Cholangiocarcinoma in the Setting of COVID-19: Unique Challenges and Opportunities
By Ghassan K. Abou-Alfa, MD, MBA; Bruce Lin, MD; Farshid Dayyani, MD, PhD; Richard Kim, MD; Rachna T. Shroff, MD, MS; Melinda Bachini; Milind M. Javle, MD
December 2020, Special Issue: Managing CCA in the Setting of COVID-19
The first confirmed case in the United States of the 2019 novel coronavirus (COVID-19) was reported on January 21, 2020, and as of November 27, 2020, there were >63 million confirmed cases and >1.5 million deaths globally. Despite the global impact, certain populations have been identified as having a higher risk of developing severe COVID-19 infection, including patients with cancer. Various studies have shown that patients with cancer experience particularly poor outcomes after COVID-19 infection. As a result of the pandemic, care delivery has been disrupted to some degree based on the need for prioritization and limitations on resources. Therefore, healthcare providers and patients have been continually reassessing the balance between the benefits and risks of anticancer interventions in the context of the added risk of COVID-19 infection.

Subscribe Today!

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

I'd like to receive: