1,825
Views
5
CrossRef citations to date
0
Altmetric
Editorial

The impact of molecular profiling on cholangiocarcinoma clinical trials and experimental drugs

& ORCID Icon
Pages 281-284 | Received 19 Sep 2020, Accepted 04 Nov 2020, Published online: 23 Nov 2020

1. Cholangiocarcinoma, molecular profiling and experimental drugs

Cholangiocarcinomas (CCAs) are a heterogeneous group of malignancies arising from the biliary tract. CCAs are categorized as intrahepatic (iCCA) and extrahepatic CCA (eCCA) depending on their anatomical site of origin. The eCCA further splits into perihilar (pCCA) and distal (dCCA) CCA [Citation1]. Heterogeneity at genomic, epigenetic, and molecular level dramatically impacts the efficacy of available systemic therapies [Citation1]. Adding to the complexity, most patients present at an advanced stage, and potentially curative resection is feasible in only 20% of the patients [Citation2]. Adjuvant capecitabine should be considered for all patients with resected biliary tract cancer, particularly those with lymph node involvement or a margin-positive resection [Citation3]. Unfortunately, most patients will either recur after potentially curative treatment or present with unresectable or metastatic disease. The standard of care for patients with unresectable biliary tract cancer is a combination of cisplatin and gemcitabine, though this combination has only demonstrated a median overall survival (mOS) of 11.7 months [Citation4]. Multiple efforts are underway to try to build on this backbone. For instance, based on promising results in a single-arm phase 2 study, SWOG 1815 is currently testing a triplet regimen of cisplatin, gemcitabine, and nab-paclitaxel (NCT03768414) [Citation5] and KHBO1401-MITSUBA study in Japan is testing a triplet regimen of (gemcitabine, cisplatin, and S-1) in first-line setting [Citation6]. Following progression first-line therapy, only 15–25% of patients can receive second-line systemic therapy depending on their performance status and organ function [Citation7]. The ABC-06 trial was the first prospective, phase III study in second-line setting, comparing FOLFOX to active symptom control. While there was a significant improvement in mOS, the results remain underwhelming [Citation8]. Most patients receiving second-line therapy have an mOS of 3–7 months [Citation7]. Immunotherapy is rarely an option as the role is limited to patients with deficient mismatch repair pathway and microsatellite instability (MSI-high). MSI-high is rare (1%), and most of these are seen in tumors harboring a high tumor mutation burden. Studies have demonstrated low PD-L1 expression (9%) and low PD-L1 amplification (0.27%) in these tumors and there is no evidence that PD-L1 expression is correlated with response to immunotherapy [Citation9].

Recent advances in next-generation sequencing (NGS) techniques and routine tumor profiling has helped define the genomic landscape of CCAs [Citation10,Citation11]. CCAs are genetically heterogeneous diseases with specific patterns based on anatomic location (iCCAs vs. eCCA), geographical location (Eastern vs. Western), and etiology (fluke, Hepatitis B, other causes) [Citation12–14]. Similarly, an integrative clustering analysis by an international collaborative effort defined four CCA clusters – Fluke-Positive CCAs (Clusters 1/2) were enriched in ERBB2 amplification and TP53 mutations, and Fluke-Negative CCAs (Clusters 3/4) exhibit high copy-number alterations and PD-1/PD-L2 expression, or epigenetic mutations (IDH1/2, BAP1) and FGFR/PRKA-related gene rearrangements [Citation15]. Somatic alterations with therapeutic implications can be found in up half of these patients [Citation11]. KRAS, SMAD4, and STK11 alterations are common in extrahepatic cholangiocarcinoma, whereas mutations in IDH1, BAP1, TP53, and FGFR2 fusions occur with greater frequency in intrahepatic cases. BAP1 mutations and FGFR2 gene fusions are exclusively in patients with intrahepatic cholangiocarcinoma [Citation11]. The evolving knowledge about the genomic profile of CCA is leading to the development of targeted therapies and supporting the use of molecular profiling to guide therapy selection in patients with advanced biliary cancers.

The MOSCATO-01 trial offered early supportive evidence (median OS 17 vs. 5 months; p = 0.008) in favor of personalized treatment of CCAs after years of negative trials involving drugs targeting pathways such as angiogenesis or the human epidermal growth factor receptor (HER [or ERBB]) family, HGF/c-MET, Hedgehog, KRAS/BRAF/MEK/ERK, and PI3K/AKT/mTOR [Citation16,Citation17]. The sea change in drug development and clinical trial activity in CCAs is revealed by a search on clinicaltrials.gov, resulting in more than 500 trials with a steady increase in trials since 2010 (). Several early phase clinical trials are either completed or underway testing drugs targeting the fibroblast growth factor receptor 2 (FGFR) fusion, isocitrate dehydrogenase (IDH) mutations, the human epidermal growth factor receptor (HER) family, chromatin remodeling genes (ARID1, BAP1, and PBRM1), DNA damage repair (DDR) pathways, BRAF, and RNF43 mutations [Citation9,Citation14,Citation17].

Figure 1. Increasing trends in registration of cholangiocarcinoma clinical trials in the last decade

Figure 1. Increasing trends in registration of cholangiocarcinoma clinical trials in the last decade

Targeting FGFR2 fusions and IDH mutations has demonstrated encouraging results () [Citation18]. In fact, targeting FGFR2 with pemigatinib, and FGFR inhibitor, is currently the only FDA approved strategy for the treatment of CCAs. FGFRs are a family of four transmembrane receptors with intracellular tyrosine kinase domains (FGFR1-4). FGFR2 tyrosine kinase fusions are seen in about 15% of iCCAs and activate multiple downstream canonical signaling events to propagate oncogenesis [Citation17,Citation19]. Hence, tyrosine kinase inhibitors (TKIs) such as ponatinib, pazopanib, regorafenib, infigratinib, derazantinib, futibatinib, and pemigatinib have been studied in several phase 1 and 2 clinical trials with encouraging results. FDA recently approved pemigatinib for the treatment of adults with previously treated, unresectable locally advanced or metastatic cholangiocarcinoma with a fibroblast growth factor receptor 2 (FGFR2) fusion or other rearrangements based on the results of FIGHT-202. Among the 107 patients, the overall responserate (ORR) was 36%, with 3 complete responses. The median duration of response (DOR) was 7.5 months with responses lasting ≥6 months in 24 of the 38 (63%) responding patients and ≥12 months in 7 (18%) patients. No responses were observed among patients with other FGF/FGFR alterations, and hence the use of pemigatinib should be currently limited to patients with FGFR2 fusions [Citation18].

Table 1. Selected published targeted therapy trials in advanced cholangiocarcinoma

Targeting IDH has also yielded promising results. The prevalence of IDH mutation is approximately 20% in patients with CCA. IDH1 mutations are more common as compared to IDH2 mutations and are mostly seen in noninfectious (Opistorchis viverrine [OV] unrelated and hepatitis-negative) iCCA. Somatic gain-of-function mutations in IDH1 and IDH2 result in the accumulation of the 2-hydroxyglutarate (2-HG), which promotes a hypermethylated phenotype and tumorigenesis by inhibiting DNA and protein demethylation [Citation20]. The recently published ClarIDHy phase III trial showed that the IDH1 inhibitor ivosidenib could lower the risk of death by 63% in CCA patients who harbor IDH1-mutations and have progressed on prior chemotherapy [Citation21]. The results of multiple ongoing trials may expand the role of IDH inhibitors beyond a limited subset of IDH-1-mutated patients, especially when IDH inhibitors are combined with chemotherapy or PARP inhibitors, as the IDH mutations can induce a homologous recombination defect similar to a ‘BRCAness’ phenotype [Citation22]. Targeting other pathways such as HER2, RAS/ERK pathway, PI3K/AKT/mTOR, DNA repair mechanisms, and BAP-1 mutations and VEGF have shown modest activity but continue to be under investigation [Citation17]. That said, impressive results were seen in BRAF mutated CCA patients in the ROAR basket trial with the dual inhibition of BRAF and MEK pathways using dabrafenib (BRAF inhibitor) and trametinib (MEK inhibitor). Here, an overall response rate of 47%, a median PFS of 9 months, and a median OS of 14 months in patients with CCA () [Citation23].

2. Expert opinion

A personalized approach to cholangiocarcinoma continues to evolve but should be considered an option for all patients with targetable alterations (). This underscores the importance of biomarker testing for every advanced CCA patient. Increasing the feasibility of conducting targeting therapy trials through international collaborations and with advances in liquid biopsy represent major advances to support personalized medicine. Several ongoing trials (the National Cancer Institute [NCI] Molecular Analysis for Therapy Choice [MATCH], GOZILA study (UMIN000029315) [Citation24] and the ASCO Targeted Agent and Profiling Utilization Registry [TAPUR] trials) are using next-generation sequencing (NGS) to identify molecular abnormalities in the tumors of patients with refractory cancers that may potentially match molecularly targeted therapies that are either in clinical trials or approved for the treatment of other cancer types. This approach can and should be leveraged in CCA too. Molecular profiling, new drug development, and availability of clinical trials have opened up the possibility of long-term survival, at least for select patients, in this devastating disease.

Figure 2. Approach to targeted therapies in cholangiocarcinoma after progression on 1st line treatment

Figure 2. Approach to targeted therapies in cholangiocarcinoma after progression on 1st line treatment

Declaration of interest

R Shroff is an Advisory Board Member for Merck, Seattle Genetics, QED Therapeutics, DebiPharm, Agios, Clovis, Exelixis, and Incyte. She also receives research funding from Merck, Haloyzme, Pieris, Taiho, Rafael Pharmaceuticals, and Exelixis Pharmaceuticals. The authors have no other 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, Lamarca A, et al. Cholangiocarcinoma 2020: the next horizon in mechanisms and management. Nat Clin Pract Gastroenterol Hepatol. 2020;17(9):557–588.
  • Shroff RT, Kennedy EB, Bachini M, et al. Adjuvant Therapy for Resected Biliary Tract Cancer: ASCO Clinical Practice Guideline. J Clin Oncol. 2019;37(12):1015–1027.
  • Primrose JN, Fox RP, Palmer DH, et al. Capecitabine compared with observation in resected biliary tract cancer (BILCAP): a randomised, controlled, multicentre, phase 3 study. Lancet Oncol. 2019;20(5):663–673.
  • Valle J, Wasan H, Palmer DH, et al. Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. N Engl J Med. 2010;362(14):1273–1281. .
  • Shroff RT, Javle MM, Xiao L, et al. Gemcitabine, cisplatin, and nab-paclitaxel for the treatment of advanced biliary tract cancers: a phase 2 clinical trial. JAMA Oncol. 2019;5(6):824–830.
  • Kawabata R, Terazawa T, Matsuyama J, et al. A multicenter phase II trial of perioperative capecitabine plus oxaliplatin for clinical stage III gastric cancer (OGSG1601). Ann Oncol. 2018;29:viii226.
  • Lamarca A, Hubner RA, David Ryder W, et al. Second-line chemotherapy in advanced biliary cancer: a systematic review. Ann Oncol. 2014;25(12):2328–2338.
  • Lamarca A, Palmer DH, Wasan HS, et al. ABC-06 | A randomised phase III, multi-centre, open-label study of active symptom control (ASC) alone or ASC with oxaliplatin/5-FU chemotherapy (ASC+mFOLFOX) for patients (pts) with locally advanced/metastatic biliary tract cancers (ABC) previously-treated with cisplatin/gemcitabine (CisGem) chemotherapy. J clin oncol. 2019;37(15_suppl):4003.
  • Tella SH, Kommalapati A, Borad MJ, et al. Second-line therapies in advanced biliary tract cancers. Lancet Oncol. 2020;21(1):e29–e41.
  • Zou S, Li J, Zhou H, et al. Mutational landscape of intrahepatic cholangiocarcinoma. Nat Commun. 2014;5:5696.
  • Lowery MA, Ptashkin R, Jordan E, et al. Comprehensive molecular profiling of intrahepatic and extrahepatic cholangiocarcinomas: potential targets for intervention. Clin Cancer Res. 2018;24(17):4154–4161.
  • Ong CK, Subimerb C, Pairojkul C, et al. Exome sequencing of liver fluke-associated cholangiocarcinoma. Nat Genet. 2012;44(6):690–693.
  • Cao J, Hu J, Liu S, et al. Intrahepatic cholangiocarcinoma: genomic heterogeneity between eastern and western patients. JCO Precis Oncol. 2020; (4):557–569. DOI:10.1200/PO.18.00414.
  • Chakrabarti S, Kamgar M, Mahipal A. Targeted therapies in advanced biliary tract cancer: an evolving paradigm. Cancers (Basel). 2020;12:8.
  • Jusakul A, Cutcutache I, Yong CH, et al. Whole-genome and epigenomic landscapes of etiologically distinct subtypes of cholangiocarcinoma. Cancer Discov. 2017;7(10):1116–1135.
  • Verlingue L, Malka D, Allorant A, et al. Precision medicine for patients with advanced biliary tract cancers: an effective strategy within the prospective MOSCATO-01 trial. Eur J Cancer. 2017;87:122–130.
  • Lamarca A, Barriuso J, McNamara MG, et al. Molecular targeted therapies: ready for “prime time” in biliary tract cancer. J Hepatol. 2020;73(1):170–185.
  • 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(5):671–684.
  • Presta M, Chiodelli P, Giacomini A, et al. Fibroblast growth factors (FGFs) in cancer: FGF traps as a new therapeutic approach. Pharmacol Ther. 2017;179:171–187.
  • Mondesir J, Willekens C, Touat M, et al. IDH1 and IDH2 mutations as novel therapeutic targets: current perspectives. J Blood Med. 2016;7:171–180.
  • 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;21(6):796–807.
  • Sulkowski PL, Corso CD, Robinson ND, et al. 2-Hydroxyglutarate produced by neomorphic IDH mutations suppresses homologous recombination and induces PARP inhibitor sensitivity. Sci Transl Med. 2017;9(375):eaal2463.
  • Subbiah V, Lassen U, Élez E, et al. Dabrafenib plus trametinib in patients with BRAF(V600E)-mutated biliary tract cancer (ROAR): a phase 2, open-label, single-arm, multicentre basket trial. Lancet Oncol. 2020;21(9):1234–1243.
  • Nakamura Y, Taniguchi H, Bando H, et al. Utility of circulating tumor DNA (ctDNA) versus tumor tissue clinical sequencing for enrolling patients (Pts) with advanced gastrointestinal (GI) cancer to matched clinical trials: SCRUM-Japan GI-SCREEN and GOZILA combined analysis. Am. J. Clin. 2020;38(4_suppl):5.

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.