1,767
Views
1
CrossRef citations to date
0
Altmetric
Editorial

Toward personalized therapy for cholangiocarcinoma: new insights and challenges

, , &
Pages 1241-1243 | Received 03 Aug 2021, Accepted 21 Sep 2021, Published online: 11 Oct 2021

1. Introduction

Cholangiocarcinoma (CCA) encompasses a group of rare and aggressive hepatobiliary malignancies, including extrahepatic cholangiocarcinoma (eCCA) and intrahepatic cholangiocarcinoma (iCCA), with the former further subdivided into perihilar (pCCA) and distal cholangiocarcinoma (dCCA) [Citation1]. Despite CCAs have been historically considered rare tumors, the incidence of CCA is on the rise in several countries, representing the second most frequently diagnosed primary liver cancer worldwide and accounting approximately for the 15–20% of all liver malignancies [Citation2]. Radical surgery is the only curative approach for CCA, but unfortunately, less than one-third of patients are diagnosed with resectable disease, and recurrence rates remain high, even following radical resection with negative tumor margins [Citation3].

More than ten years ago, the Advanced Biliary tract Cancer (ABC)-02 phase III trial established the combination of cisplatin plus gemcitabine (CisGem) as the standard of care in the front-line setting [Citation4]; more recently, the ABC-06 trial highlighted the survival benefit provided by modified oxaliplatin plus 5-fluorouracil (mFOLFOX) plus active symptom control (ASC) over ASC alone in CCA patients whose disease progressed on first-line CisGem [Citation5]. However, systemic chemotherapy is marginally effective in locally advanced/unresectable or metastatic CCA, being associated with a median overall survival (mOS) shorter than 1 year.

As previously stated, the term CCA includes a heterogeneous group of malignancies with important anatomical, epidemiological, and molecular differences [Citation6]. As regards the latter, several types of genetic aberrations have been observed in CCA, ranging from single-nucleotide mutations (such as missense, nonsense, and splice-site mutations) from insertions and deletions (e.g. BAP1, TP53, ARID1A) as well as copy number alterations and chromosomal rearrangements or fusions (e.g. FGFR2, ALK, ROS1, and NTRK) [Citation7]. Of note, these genetic aberrations cluster in specific CCA subgroups: IDH1 mutations and FGFR2 fusions are the most frequent actionable alterations in iCCAs, while ERBB2 amplifications and ARID1A and PIK3CA mutations are more common in eCCA patients [Citation8].

In the current Editorial, we provide a critical overview of personalized therapies in CCA management, especially focusing on current and future challenges in this setting.

2. Expert opinion

Certainly, not all CCAs are suitable for molecularly driven treatments, and several issues are to be faced, including the quality of tissue sample [Citation9]. We know from everyday clinical practice that the pathologic confirmation of diagnosis is necessary before any non-surgical treatment and may be challenging in CCA, especially in patients affected by primary sclerosing cholangitis and biliary strictures. In addition, despite endoscopic imaging and tissue sampling are useful, biopsy samples are often inadequate for molecular profiling, and the highly desmoplastic nature of CCA limits the accuracy of cytological and pathological approaches [Citation10]. Based on these premises, these issues have remarked the urgent need to develop novel strategies and methodologies able to anticipate the CCA diagnosis at an early stage, as well as to obtain sufficient material to perform genomic analysis. Among these strategies, liquid biopsy has received growing attention in several malignancies, including CCA [Citation11]. In fact, liquid biopsy has shown high concordance with tissue biopsy, representing a noninvasive method able to capture heterogeneous resistance alterations. However, these methodologies are still preliminary and quite far from everyday clinical practice for now.

Targeted therapies have provided no benefit in ‘non-selected’ CCA, and since approximately 60% of all CCAs are not suitable for targeted therapies, only a relatively small and specific population of CCA patients may benefit from these approaches [Citation12]. As regards clinical trials design, the CCA medical community has proposed three main different approaches aimed to evaluate targeted therapies in these hepatobiliary malignancies: 1) target-specific clinical trials on CCA patients (e.g. IDH1 mutations, FGFR2 fusions); 2) target-specific basket trials including a CCA cohort (e.g. NTRK fusions, BRAF mutations, MMR deficiency and other disease-agnostic aberrations); 3) CCA umbrella or basket trials with target-specific arms. As regards the former type of studies on CCA, several trials on specific actionable targets have been presented and published in the last years. A key example is FGFR2 fusions, with these genetic aberrations representing an important molecular driver in iCCA, as witnessed by the Food and Drug Administration (FDA) approval of infigratinib and pemigatinib and the large number of FGFR-targeted therapies under evaluation in this setting [Citation13–15].

In the ROAR phase II basket trial, the combination of dabrafenib plus trametinib has reported a promising activity and a manageable safety profile in the cohort of patients with BRAFV600E-mutated CCA [Citation16]. In particular, median progression-free survival (PFS) and median overall survival (OS) were 9 months and 14 months, respectively, in CCA patients receiving dabrafenib plus trametinib, with a median duration of response of 9 months and an overall response rate of 47% [Citation16]. Lastly, as regards umbrella trials with target-specific arms, the SAFIR-ABC10 study will see the evaluation of multiple targeted therapies for CCA patients that will be stratified into multiple subgroups based on molecular features screened by FoundationOne testing.

In addition, immunotherapy is currently under assessment in advanced CCA, as monotherapy or in combination with targeted therapies, including targeted therapies and tyrosine kinase inhibitors including lenvatinib [Citation17]. In terms of monotherapy, data from CCA patients treated with pembrolizumab in the phase Ib KEYNOTE-028 and the phase II KEYNOTE-158 trials have been recently published [Citation18,Citation19]. As regards the former, monotherapy with the PD-1 inhibitor pembrolizumab (10 mg/kg every 2 weeks) has been evaluated in previously treated patients with PD-L1 positive metastatic solid malignancies, including 20 CCAs and 4 gallbladder cancers [Citation18]. After a median follow-up of 5.7 months, overall response rate (ORR) was 13%, with median OS and PFS of 5.7 and 1.8 months, respectively [Citation18]. MSI status was not available in approximately 30% of CCA patients enrolled in this study. Similarly, the KEYNOTE-158 basket trial evaluated single-agent pembrolizumab as second- or later-line treatment in advanced solid tumors, including 104 biliary tract cancers [Citation18,Citation19], where the PD-1 inhibitor reported a disappointing ORR of 5.8% [Citation18]. Median PFS and OS were 2.0 (95% CI, 1.9–2.1) and 7.4 (95% CI, 5.5–9.6) months, respectively. Of note, KEYNOTE-028 and KEYNOTE-158 enrolled patients showing disease progression following at least 1 prior systemic therapy, many of whom were heavily pretreated. In 2017, the US Food and Drug Administration approved the anti-PD-1 agent pembrolizumab for the treatment of any dMMR or MSI-H malignancies, regardless of tumor type, including CCA; in addition, in June 2020 the PD-1 inhibitor pembrolizumab was approved by the FDA also for TMB high (more than 10 mutations per megabase) non-colorectal malignancies. Thus, these agents are currently used for CCA with putative predictive biomarkers.

In summary, molecularly targeted therapies are providing CCA patients new treatment options, and we are witnessing important efforts aiming to expand the role of other emerging agents as well as to evaluate novel strategies able to overcome drug resistance [Citation20]. We have recently seen important changes in the treatment paradigm of CCA, and the close collaboration between clinicians and researchers is and will be of great importance to improve clinical outcomes of CCA patients [Citation21]. On the horizon, there is a host of phase II and phase III trials of novel treatments, including studies comparing targeted therapies versus standard chemotherapy in the front-line setting [Citation22,Citation23]; the results of these trials are highly awaited and have the potential to modify the treatment landscape of these challenging hepatobiliary malignancies with many unanswered questions.

Declaration of interests

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Additional information

Funding

This paper was not funded.

References

  • Banales JM, Marin JJG, and Lamarca A, et al. Cholangiocarcinoma 2020: the next horizon in mechanisms and management. Nat Rev Gastroenterol Hepatol. 2020;17(9) :557-588. DOI:https://doi.org/10.1038/s41575-020-0310-z.
  • Saha SK, Zhu AX, Fuchs CS, et al. Forty-year trends in cholangiocarcinoma incidence in the US: intrahepatic disease on the rise. Oncologist. 2016;21:594–599.
  • Rizvi S, Khan SA, Hallemeier CL, et al. Cholangiocarcinoma ‐ evolving concepts and therapeutic strategies. Nat Rev Clin Oncol. 2018;15(2):95–111.
  • Valle J, Wasan H, Palmer DH, et al.; ABC-02 Trial Investigators. Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. N Engl J Med. 2010;362:1273–1281.
  • Lamarca A, Palmer DH, Wasan HS, et al.; Advanced Biliary Cancer Working Group. Second-line FOLFOX chemotherapy versus active symptom control for advanced biliary tract cancer (ABC-06): a phase 3, open-label, randomised, controlled trial. Lancet Oncol. 2021 May;22(5):690–701.
  • Forner A, Vidili G, Rengo M, et al. Clinical presentation, diagnosis and staging of cholangiocarcinoma. Liver Int. 2019;39(Suppl 1):98–107.
  • Kelley RK, Bridgewater J, Gores GJ, et al. Systemic therapies for intrahepatic cholangiocarcinoma. J Hepatol. 2020;72(2):353–363.
  • Abou-Alfa GK, Macarulla T, Javle MM, et al. Ivosidenib in IDH1-mutant, chemotherapy-refractory cholangiocarcinoma (ClarIDHy): a multicentre, randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol. 2020 Jun;21(6):796–807. Epub 2020 May 13. Erratum in: Lancet Oncol. 2020 Oct;21(10):e462.PMID: 32416072; PMCID: PMC7523268.
  • Rizzo A, Frega G, Ricci AD, et al. Anti-EGFR monoclonal antibodies in advanced biliary tract cancer: a systematic review and meta-analysis. In Vivo. 2020Mar-Apr;34(2):479–488. PMID: 32111744; PMCID: PMC7157865.
  • Lang SA, Bednarsch J, Joechle K, et al. Prognostic biomarkers for cholangiocarcinoma (CCA): state of the art. Expert Rev Gastroenterol Hepatol. 2021May;15(5):497–510. Epub 2021 May 10. PMID: 33970740.
  • Goyal L, Shi L, Liu LY, et al. TAS-120 overcomes resistance to ATP-competitive FGFR inhibitors in patients with FGFR2 fusion-positive intrahepatic cholangiocarcinoma. Cancer Discov. 2019 Aug;9(8):1064–1079. https://doi.org/10.1158/2159-8290.CD-19-0182. Epub 2019 May 20. PMID: 31109923; PMCID: PMC6677584.
  • Mahipal A, Kommalapati A, Tella SH, et al. Novel targeted treatment options for advanced cholangiocarcinoma. Expert Opin Investig Drugs. 2018Sep;27(9):709–720. Epub 2018 Aug 30. PMID: 30124336.
  • Abou-Alfa GK, Sahai V, Hollebecque A, et al. Pemigatinib for previously treated, locally advanced or metastatic cholangiocarcinoma: a multicentre, open-label, phase 2 study. Lancet Oncol. 2020;21:671–684.
  • Rizzo A, Ricci AD, Brandi G. Futibatinib, an investigational agent for the treatment of intrahepatic cholangiocarcinoma: evidence to date and future perspectives. Expert Opin Investig Drugs. 2020 Oct;25:1–8.
  • Zhang W, Zhou H, Wang Y, et al. Systemic treatment of advanced or recurrent biliary tract cancer. Biosci Trends. 2020 Nov 4;14(5):328–341. Epub 2020 Aug 24. PMID: 32830166.
  • Subbiah V, Lassen U, Élez E, et al. Dabrafenib plus trametinib in patients with BRAFV600E-mutated biliary tract cancer (ROAR): a phase 2, open-label, single-arm, multicentre basket trial. Lancet Oncol. 2020Sep;21(9):1234–1243. Epub 2020 Aug 17. PMID: 32818466.
  • Lin J, Yang X, Long J, et al. Pembrolizumab combined with lenvatinib as non-first-line therapy in patients with refractory biliary tract carcinoma. Hepatobiliary Surg Nutr. 2020Aug9;9(4):414–424. PMID: 32832493; PMCID: PMC7423565.
  • 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 Oct 15;147(8):2190–2198.
  • Rizzo A, Ricci AD, Brandi G. Recent advances of immunotherapy for biliary tract cancer. Expert Rev Gastroenterol Hepatol. 2021May;15(5):527–536. Epub 2021 Jan 8. PMID: 33215952.
  • Sipra QUAR, Shroff R. The impact of molecular profiling on cholangiocarcinoma clinical trials and experimental drugs. Expert Opin Investig Drugs. 2021Apr;30(4):281–284. Epub 2020 Nov 23. PMID: 33228417.
  • Wang J, Ilyas S. Targeting the tumor microenvironment in cholangiocarcinoma: implications for therapy. Expert Opin Investig Drugs. 2021Apr;30(4):429–438. Epub 2020 Dec 28. PMID: 33322977; PMCID: PMC8096665.
  • Rahnemai-Azar AA, Weisbrod AB, Dillhoff M, et al. Intrahepatic cholangiocarcinoma: current management and emerging therapies. Expert Rev Gastroenterol Hepatol. 2017May;11(5):439–449. Epub 2017 Mar 29. PMID: 28317403.
  • Okusaka T. Cholangiocarcinoma: is it time for a revolution? Expert Rev Gastroenterol Hepatol. 2021May15;15(5):467–470. Epub 2021 Apr 15. PMID: 33840344.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.