Recent advancements in systemic therapy for biliary tract cancers: a literature review
Introduction
Biliary tract cancers (BTCs) are a diverse and anatomically complex group of malignancies arising from the epithelial lining of the biliary system and include gallbladder cancer (GBC), intrahepatic cholangiocarcinoma (iCCA), perihilar cholangiocarcinoma (Klatskin tumors), extrahepatic cholangiocarcinoma (eCCA), and, occasionally, ampullary cancers (1,2). Among these, GBC is the most common subtype, followed by eCCA and iCCA; however, precision in epidemiology has been historically hindered by the frequent misclassification of perihilar tumors as iCCAs (3,4). BTCs impose a substantial global health burden and were responsible for more than 211,000 new diagnoses and approximately 174,000 deaths worldwide in 2017 (5). Although BTCs are considered rare on a global scale, their incidence has been rising steadily over recent decades, with Southeast Asia reporting the highest regional burden (6,7). Incidence rates vary dramatically by geography, ranging from 0.3 to 6 per 100,000 individuals; for example, in South Korea, the reported rate is 3.0 per 100,000, while in the United Kingdom, it is 0.66 per 100,000 (8). Despite constituting only about 15% of all primary liver tumors, BTCs are notoriously aggressive and confer a poor prognosis, with the 5-year overall survival (OS) rate remaining below 10% and the recurrence rate exceeding 50% even among early-stage cases (9-11). Numerous risk factors have been implicated in the pathogenesis of BTCs, including hepatobiliary infections, gallstones, liver cirrhosis, diabetes mellitus, obesity, chronic hepatitis B and C infections, and, notably, primary sclerosing cholangitis (PSC), which is particularly prevalent in Asian populations (9). Due to nonspecific symptoms and inherent biological heterogeneity, the majority of patients with BTCs are diagnosed at advanced or metastatic stages, limiting the opportunity for systemic treatment, participation in clinical trials, or palliative care (12). In this challenging context, the identification of reliable prognostic and predictive biomarkers is essential for guiding personalized treatment approaches. Inflammatory indices derived from routine blood parameters such as the neutrophil-to-lymphocyte ratio (NLR) and the emerging neutrophil-to-eosinophil ratio (NER) have demonstrated significant prognostic relevance across various malignancies, including BTCs (13). The Royal Marsden Hospital (RMH) prognostic score, although originally developed for patients in early-phase clinical trials, has proven useful in BTC cohorts. It integrates serum albumin levels, lactate dehydrogenase (LDH), and the number of metastatic sites to stratify patients according to risk and assist with trial enrollment and treatment planning (14). Molecular profiling has identified several actionable genetic alterations in BTCs. Up to 15–20% of iCCA tumors harbor isocitrate dehydrogenase 1 (IDH1) mutations or fusions involving the fibroblast growth factor receptor (FGFR) family, both of which are being actively targeted in ongoing clinical trials. Additionally, B-Raf proto-oncogene, serine/threonine kinase (BRAF) V600E mutations are found in approximately 5% of iCCA cases and represent a promising target for genotype-directed therapies involving dual BRAF and MEK inhibition (15). Although human epidermal growth factor receptor 2 (HER2) amplification or overexpression is rare in iCCA, it is observed in up to 15–20% of GBCs and eCCAs, which forms the rationale for anti-HER2 therapeutic strategies in these subsets (15-17). Support for the targeting of BRAF is primarily derived from a phase II multicenter basket trial, which demonstrated the clinical benefit of combined BRAF-MAPK/ERK pathway inhibition in patients with BRAF V600E-mutated tumors. Approximately 5% of iCCA cases carry this activating mutation in the BRAF gene, which drives tumorigenesis through the MAPK pathway (18). HER2 activation is also present in a subset of BTC cases. Although rare in iCCA, HER2 overexpression or gene amplification is present in 15–20% of eCCA and GBC cases, providing further justification for the application of HER2-targeted therapy in these populations (19). However, HER2 alterations are relatively uncommon in BTCs overall, occurring in just 1–2% of cases. In contrast, they are more prevalent in ampullary cancers, with reported rates of up to 7% (20). Alterations in neurotrophic receptor tyrosine kinase (NTRK) genes 1–3 are exceedingly rare in BTCs but are considered actionable targets when present, given the efficacy of NTRK inhibitors in fusion-positive cancers (21). Similarly, a small subset of patients with BTC, approximately 1% harbor high microsatellite instability (MSI-H) or mismatch repair deficiency (dMMR), forming a rare but therapeutically relevant population that may benefit from immunotherapy (22). Given the predominance of late-stage diagnoses, BTCs are often associated with poor clinical outcomes, and current treatment strategies are largely limited to systemic therapies, investigational agents in clinical trials, or supportive care (12).
The therapeutic approaches that are presently in use or that are being considered by clinicians for the systemic treatment of BTCs are summarized in the sections below. We present this article in accordance with the Narrative Review reporting checklist (available at https://jgo.amegroups.com/article/view/10.21037/jgo-2025-83/rc).
Methods
Google Scholar, PubMed, EMBASE, and the Cochrane Library were searched for recent literature via the following search terms: bile duct cancer, systemic therapy, adjuvant therapy, chemotherapy, cholangiocarcinoma, extrahepatic cholangiocarcinoma, intrahepatic cholangiocarcinoma, distal cholangiocarcinoma, common bile duct cancer, gallbladder cancer, targeted therapy, immunotherapy, and similar terms.
Literature selection
The inclusion criteria for the literature were as follows: (I) English-language literature; (II) extensive studies; (III) a topic related to recent advancements in BTCs, and (IV) publication in peer-reviewed journals. Meanwhile, the exclusion criteria for the literature were as follows: (I) inadequate data for BTCs; (II) similar studies from the same institute or sharing the same patient sample; and (III) languages other than English (see Table 1).
Table 1
| Items | Specification |
|---|---|
| Date of search | January 1, 2025 |
| Databases and other sources searched | PubMed, Google Scholar, EMBASE, and the Cochrane Library |
| Search terms used | bile duct cancer, systemic therapy, adjuvant therapy, chemotherapy, cholangiocarcinoma, extrahepatic cholangiocarcinoma, intrahepatic cholangiocarcinoma, distal cholangiocarcinoma, common bile duct cancer, gallbladder cancer, targeted therapy, immunotherapy |
| Timeframe | December 2010 to December 2024 |
| Inclusion criteria | English-language literature including randomized controlled trials, recent original studies, and systematic reviews and meta-analyses |
| Exclusion criteria | (I) Studies with inadequate data for BTCs; (II) similar studies from the same institute or that used the same patient sample; (III) languages other than English |
| Selection process | M.M.H. conducted the literature search and identified the eligible literature. Consensus was obtained via discussion among all the authors and corresponding authors |
BTC, biliary tract cancer.
Treatment
Systemic treatments in the advanced setting are based on three main pillars: cytotoxic chemotherapy, the addition of immunotherapy to chemotherapy, and targeted therapies. Targeted therapies are based on several promising targets, including IDH1 mutations, FGFR2 fusions, BRAF-V600E mutations, and HER2 amplifications (23).
Adjuvant therapies
Adjuvant therapy represents one of the most effective approaches in improving patients’ outcomes in systemic therapy. However, several issues exist, and the results from trials are equivocal, most likely due to the diversity of BTCs observed in terms of prognosis, etiology, epidemiology, and molecular characteristics. The only effective treatment available remains radical surgical resection; unfortunately, however, only 35% patients at diagnosis have resectable disease, and postoperative recurrence is common (50–70%) (24-27). Therefore, there is a critical need to develop effective adjuvant therapy. In the PRODIGE 12 ACCORD 18 clinical trial and BCAT trial (28), two recent observation-controlled phase III trials, adjuvant gemcitabine-based chemotherapy was not found to be effective in treating surgically removed biliary tract malignancies (29). Perioperative chemotherapy may thus be necessary to improve survival, but as BTCs are relatively uncommon, few randomized phase III clinical study trials on perioperative chemotherapy have been conducted. For advanced BTCs, gemcitabine combined with cisplatin plus S-1 (GCS) has become the preferred chemotherapeutic regimen. In addition to increasing OS, GCS is associated with a high response rate. A phase III randomized clinical trial (JCOG1920) conducted in Japan examined the effectiveness of GCS in preoperative neoadjuvant chemotherapy for resectable BTCs (30), yet clinical studies that have assessed the value of adjuvant systemic treatment for patients with BTCs are rare (Tables 2,3).
Table 2
| Study | N | Intervention | Outcomes |
|---|---|---|---|
| BEER BTC (31), CTRI/2017/10/010112 | 98 | Bevacizumab and erlotinib | Patients in the BEER cohort had better outcomes than those in the control arm, with a median PFS of 5.2 months (95% CI: 3.4–6.9) versus 3.0 months (95% CI: 2.5–3.5; P=0.004). By tumor type, GBC patients in the BEER arm had a median OS of 4.3 vs. 3.0 months in controls, while CC patients showed a larger difference (6.7 vs. 2.2 months) |
| (BilT-04) (32), NCT04203160 | 20 | Combination of devimistat with cisplatin and gemcitabine | The ORR was 45% and the median PFS was 10 months (95% CI: 7.1–14.9), after a median follow-up of 15.6 months. The average OS has not been determined. For patients with untreated advanced BTCs, devimistat in combination with GC was well-tolerated and had a satisfactory safety profile |
| BILCAP study (25), NCT00363584 | 447 | Capecitabine | The safety margin was 0.84 (95% CI: 0.67–1.06), with a median OS of 49.6 months (95% CI: 35.1–59.1) in the capecitabine group and a median OS of 36.1 months (95% CI: 29.7–40.2) in the control group |
| PRODIGE 12 (28), EudraCT 2008-004560-39. | 196 | Monitoring or GEMOX | The median OS in the treatment arm and control arm was 75.8 months and 50.8 months, respectively (HR =1.08, 95% CI: 0.70–1.66; P=0.74). The median RFS in the treatment arm and control arm was 30.4 and 18.5 months, respectively (HR =0.88, 95% CI: 0.62–1.25; P=0.48) |
BEER, bevacizumab + erlotinib; BTC, biliary tract cancer; CC, cholangiocarcinoma; CI, confidence interval; CTRI, Clinical Trials Registry – India; GBC, gallbladder cancer; GC, gemcitabine and cisplatin; GEMOX, gemcitabine and oxaliplatin; HR, hazard ratio; N, number of patients; NCT, National Clinical Trial; ORR, objective response rate; OS, overall survival; PFS, progression-free survival; RFS, recurrence-free survival.
Table 3
| Clinical trial | Country | Phase | Estimated enrollment, n | Targeted tumor subtypes | Interventions and arms | Primary endpoints | Secondary endpoints | Estimated completion date/status |
|---|---|---|---|---|---|---|---|---|
| Adjuvant setting | ||||||||
| ACTICCA-1 (NCT02170090) | Germany (66 study sites) | Phase III randomized open-label | 781 | GBC, CCA | Experimental arm: GC; comparison arm: capecitabine | DFS | Tolerability, safety, QoL, RFS, and OS | April 2027 |
| Gemcitabine + capecitabine vs. capecitabine in curatively resected biliary tract cancer (NCT04401709) | Korea | Phase III randomized open-label | 490 | BTC | Experimental arm: gemcitabine plus capecitabine; comparison arm: capecitabine | DFS | OS | March 2025 |
| AdBTC-1 (NCT03779035) | China | Phase III randomized open-label | 460 | GBC and CCA | Experimental arm: gemcitabine plus capecitabine; comparison arm: capecitabine | DFS | OS | December 2023 |
| NCT02548195 | China | Phase III | 286 | iCCA | Cisplatin and gemcitabine vs. capecitabine | DFS | N/A | Unknown |
| NCT02798510 | China | Phase III | 140 | pCCA, dCCA, GBC | Gemcitabine/capecitabine → gemcitabine/capecitabine vs. gemcitabine/capecitabine → CRT (capecitabine) | 2-year OS | N/A | Unknown |
| EudraCT (2010-020480-21) | Germany | Phase II | 45 | iCCA | Gemcitabine post–liver transplantation | Completion rate | Recruiting | |
| ACCORD (NCT04333927) |
China | Phase II | 92 | CCA, GBC | Camrelizumab + CRT (capecitabine) vs. surveillance | 2-year OS | June 2024 | |
| NCT04295317 | China | Phase II | 65 | iCCA | Capecitabine with anti-PD-L1 (SHR-1210) | 2-year DFS | February 2024 | |
| NCT04077983 | China | Phase II | 40 | iCCA | Gemcitabine with nab-paclitaxel | DFS | Not yet recruiting | |
| NCT04782804 | China | Phase I–II | 30 | iCCA | Tislelizumab and capecitabine | DFS | May 2024 | |
| Neoadjuvant setting | ||||||||
| GAIN (NCT03673072) | Germany | Phase III randomized open-label | 300 | GBC and BTC | Arm comparison: standard of care, no prior chemotherapy Experimental arm: neoadjuvant and GC adjuvant chemotherapy |
OS | QoL, PFS and, R0 resection rates | November 2024 |
| OPTIC (NCT05514912) | USA | Phase II non randomized | 40 | Intrahepatic CCA | Experimental arm A: infigratinib, GC, and nab-paclitaxel for FGFR2 fusion/translocation-positive patients; active comparator arm B: nab-paclitaxel with GC in FGFR2 fusion/translocation-negative patients | Tolerability, safety, and feasibility | OS, RECIST, ctDNA, RFS | June 2027 |
| POLCAGB (NCT02867865) | India | Phase II/III randomized open-label | 314 | GBC | Experimental arm: neoadjuvant CRT (45–55 Gy/25 fractions and GC); comparison arm: adjuvant GC | OS | QoL, resection rate, PFS | September 2027 |
| NCT03603834 | Thailand | Phase II | 25 | CCA | mFOLFOXIRI | ORR | December 2023 | |
| NCT04308174 (DEBATE) | Korea | Phase II | 45 | CCA, GBC | Gemcitabine + cisplatin vs. cisplatin + durvalumab | R0 rate | December 2024 | |
| NCT04669496 | China | Phase II–III | 178 | iCCA with high-risk recurrence features | Adjuvant capecitabine after surgery following gemcitabine + oxaliplatin + lenvatinib + toripalimab | Event-free survival | Recruiting | |
| NCT04559139 | USA | Phase II–III | 186 | Incidental GBC | +/− revision surgery → adjuvant cisplatin + gemcitabine → neoadjuvant cisplatin + gemcitabine | 5-year OS | Recruiting June 2029 |
|
| NCT04378023 | Spain | Phase IV | 34 | pCCA | Until transplant, EBRT + capecitabine → cisplatin + gemcitabine | 1-, 3-, and 5-year OS | Recruiting | |
| NCT04523402 | China | Phase II | 100 | iCCA with high-risk LN metastases | Gemcitabine and oxaliplatin | 24-month event-free survival | Unknown status | |
Features of LN at high risk include tumor size greater than 5 cm, numerous tumor nodules, vascular invasion, and metastases to hilar lymph nodes. This table was adapted from an open access article (33) under the terms of the Creative Commons CC BY license. BTC, biliary tract cancer; CCA, cholangiocarcinoma; CRT, chemoradiotherapy; ctDNA, circulating tumor DNA; dCCA, distal cholangiocarcinoma; DFS, disease-free survival; EBRT, external beam radiotherapy; FGFR, fibroblast growth factor receptor; GBC, gallbladder cancer; GC, gemcitabine and cisplatin; iCCA, intrahepatic cholangiocarcinoma; LN, lymph node; mFOLFOXIRI, fluorouracil + oxaliplatin + irinotecan; N/A, not applicable; ORR, objective response rate; OS, overall survival; pCCA, perihilar cholangiocarcinoma; PD-L1, programmed cell death ligand 1; PFS, progression-free survival; QoL, quality of life; RECIST, Response Evaluation Criteria in Solid Tumors; RFS, recurrence-free survival.
Depending on these findings, the American Society of Clinical Oncology (ASCO) guidelines recommend adjuvant capecitabine for approximately 6 months after curative surgical resection of BTCs, including CCA and GBC (34). Clinicians agree that after surgery, patients should recover for 12 to 16 weeks before beginning optimal therapy. In addition, chemotherapy should be administered for a limited time (rather than waiting until the disease relapses) because it is considered a routine practice in optimal conditions and has been observed to be safe. Finally, fluoropyrimidine-based therapy seems to be preferred (26).
Palliative therapy
Chemotherapy remains the cornerstone of treatment for most newly diagnosed patients with advanced BTCs, including those with unresectable or metastatic disease. Cytotoxic drugs have been observed to be effective in a number of randomized controlled trials in patients with advanced BTCs. Combination chemotherapy is currently recommended for patients with sufficient organ function and good performance status, as it is generally associated with superior responsiveness, OS, and progression-free survival (PFS) than is single-agent chemotherapy (35).
First-line palliative treatment
Standard first-line chemotherapy with cisplatin and gemcitabine has been practiced for over the past 10 years, with the ABC 02 study in the United Kingdom reporting a median survival of 11.7 months and a PFS of 8.0 months under this regimen (36). Another phase II study (37) examined the combination of gemcitabine, cisplatin, and nab-paclitaxel (GCNP) in patients with BTCs and found it conferred a higher response rate and longer survival. From January 2018 to August 2022, patients with locally advanced, inoperable, and metastatic liver disease receiving first-line chemotherapy for GCNP at a multidisciplinary joint clinic (MDJC) were evaluated, with the objective response rate (ORR) being the primary endpoint and event-free survival (EFS) being the secondary endpoint. A total of 142 patients received GCNP at baseline. Men accounted for 81.7% and women for 18.3% of the cohort, with a mean age of 52 years (range, 21–79 years). A total of 137 patients were included in the response rate calculation, with 9 (6.3%), 87 (61.3%), and 24 (16.9%) patients exhibiting CR, partial response, and stable disease, respectively, representing an ORR of 67.6% and a clinical benefit rate of 84.5%. A median EFS of 9.92 [95% confidence interval (CI): 7.69–12.14] per month was achieved. Surgery was performed in 17 of 52 (34%) patients, with neoadjuvant chemotherapy-targeted GCNP being administered for locally advanced GBC (37). The efficacy of folinic acid plus fluorouracil (5-FU) and gemcitabine plus oxaliplatin (GEMOX) versus that of best supportive care (BSC) was evaluated in 81 patients with advanced GBC. The median OS was 9.5 months for patients receiving GEMOX, 4.6 months for those treated with 5-FU plus folinic acid, and 4.5 months for those treated with BSC (P=0.039) (38).
The TOPAZ-1 trial demonstrated that compared to GC alone, durvalumab with GC provides markedly longer OS [12.8 vs. 11.5 months; hazard ratio (HR) =0.80, 95% CI: 0.66–0.97; P=0.021] (39). These OS results support the National Comprehensive Cancer Network Guidelines (NCCN) 2022 (40) recommendation that durvalumab with GC be used as one of the first-line treatments for metastatic and incurable BTC. These immune checkpoint inhibitors (ICIs), however, target programmed cell death ligand 1 (PD-L1), and have a distinct safety profile and efficacy as compared to ICIs targeting programmed cell death protein 1 (PD-1) (41). A 2019 Japanese phase III trial included 354 patients with advanced BTC and examined the noninferiority of gemcitabine plus S-1 (GS) to oral fluoropyrimidine with gemcitabine and cisplatin (GC). GS was found to be noninferior to GC. The response rate was 29.8% with GS and 32.4% with GC. The median PFS was 6.8 months for the GS group and 5.8 months for the GC group (HR =0.86, 95% CI: 0.70–1.07), while the median OS was 15.1 months for the GS group and 13.4 months for the GC group (HR =0.94, 95% CI: 0.78–1.15; P=0.046 for noninferiority). However, North America and Europe currently do not have access to S-1 (42). Another common regimen for treating BTC is GEMOX, which replaces cisplatin and oxaliplatin when combined with gemcitabine. GEMOX was evaluated in a phase II trial (43) for patients with advanced BTC not receiving chemotherapy. The results showed that the median PFS ranged from 3.4 to 5.7 months, the median OS ranged from 8.8 to 15.4 months, and the ORR ranged from 14.9% to 41% (42,44-47). There are only a few phase III clinical trials (Table 4) that have evaluated the efficacy of chemotherapy for patients with advanced BTC.
Table 4
| Study | Number of patients | Treatment | Intervention | Outcomes |
|---|---|---|---|---|
| ABC 02 Trial (36), (NCT00262769) | 410 | First line | Gemcitabine alone itself or GC | Median PFS was 5.0 months with gemcitabine and 8.0 months with GC (P<0.001). The median OS was 11.7 months in the GC group and 8.1 months in the gemcitabine group (P<0.001) |
| FUGA-BT trial (42), (UMIN000010667) | 320 | First line | GS or GC | The PFS was 5.8 months with GC and 6.8 months with GS. The median noninferiority OS was 15.1 months with GS and 13.4 months with GC |
| Kim et al. (48) (NCT01470443) | 222 | First line | GEMOX or XELOX | The PFS was 5.8 months for XELOX and 5.3 months for GEMOX. The median OS was 10.6 months for XELOX and 10.4 months for GEMOX (P=NS) |
| Sakai et al. (49), (NCT02182778) | 246 | First line | GC or GC and S-1 | The PFS was 7.4 months with GC and S-1 and 5.5 months with GC (P=0.0015). The median OS was 12.6 months for GC and 13.5 months for GC and S-1 (P=0.046) |
GC, gemcitabine and cisplatin; GEMOX, gemcitabine and oxaliplatin; GS, gemcitabine and S-1; NS, not significant; OS, overall survival; PFS, progression-free survival; XELOX, capecitabine and oxaliplatin.
KEYNOTE-966, a double-blind, placebo-controlled, randomized phase III trial, was conducted in 175 medical institutions across the world. Eligible individuals were required to be least 18 years of age and have untreated, incurable, locally advanced, or metastatic lung disease. Between October 4, 2019 and June 8, 2021, 1,564 patients were examined, of whom 1,069 were randomized to receive GC (n=536) or pembrolizumab plus GC (n=533). The median follow-up at the time of the final analysis was 25.6 months (IQR, 21.7–30.4 months). The pembrolizumab group had a median OS of 12.7 months (95% CI: 11.5–13.6), and the placebo group had a median OS of 10.9 months (IQR, 9.9–11.6 months) (HR =0.83, 95% CI: 0.72–0.95; one-sided P=0.003; limit-of-significance P=0.020). The findings indicated that pembrolizumab in combination with GC is a promising new treatment option for patients with metastatic or unresectable BTC (50). A number of clinical investigations on GTC are still in progress, and further developments are anticipated.
Second-line palliative treatment
A few second-line therapeutic options are available for patients with advanced BTC (51). However, there has been a lack of definitive findings indicating that second-line chemotherapy increases survival in those with advanced BTC. A review of 25 publications, most of which were phase II trials published before 2015, and included a total of 761 patients with advanced BTC who had progressed on first-line chemotherapy, concluded that there was insufficient evidence to support a general recommendation for second-line therapy in this setting (52). However, the efficacy of second-line treatment may vary depending on the tumor’s mutational profile. For patients without contraindications to targeted therapies and without actionable mutations, chemotherapy remains the preferred option (53).
The present standard of care for patients with advanced BTCs is second-line therapy with 5-FU plus oxaliplatin (FOLFOX) after first-line GC. The ABC 06 trial on patients progressing after GC showed that FOLFOX improved OS against active symptom control alone (median OS: 6.2 vs. 5.3 months; P=0.031; HR =0.69, 95% CI: 0.50–0.97); this advantage was also seen at 6 months (35.5% vs. 50.6%) and 12 months (11.4% vs. 25.9%). Recent results from phase II of the NIFTY trial (45) comparing patients treated with 5-FU and nanoliposomal irinotecan (nal-IRI) to those with 5-FU alone who have progressed after receiving GC therapy show promise. The median PFS in the combination arm was 7.1 months, while that in the 5-FU-alone arm was 1.4 months (HR =0.56; 95% CI: 0.39–0.81; P=0.0019), suggesting that the actual goal of the study has been met. Early-phase data from the ongoing phase III SWOG S1815 clinical trial indicated increases in median PFS and OS from the combination of GC with nab-paclitaxel used in first-line treatment (54). The NIFTY trial demonstrated that second-line treatment with nanoliposomal irinotecan combined with 5-FU and leucovorin significantly improved PFS compared to 5-FU and leucovorin alone in patients who had progressed after first-line gemcitabine-cisplatin (55). The NapaSinti trial (56), a prospective, randomized, open-label, phase II study, was recently conducted at a tertiary care hospital in Chengdu, China. It evaluated the safety and efficacy of sintilimab plus nab-paclitaxel as second-line therapy for advanced BTC. The ongoing Brightline-2 clinical trial is examining the safety and efficacy of brigimadlin (also known as BI 907828), an experimental medication, in patients with advanced or metastatic malignancies. This trial aims to evaluate brigimadlin’s potential as a novel therapy option for individuals with lung adenocarcinoma, pancreatic ductal adenocarcinoma (PDAC), bladder cancer, or advanced BTC (34). Nonetheless, only a few significant trials (Table 5) on second-line treatment in patients with advanced BTC have been completed.
Table 5
| Clinical trial | Country | Phase and design | Estimated enrollment, n | Intervention | Primary endpoint | Outcome | Completion date/estimated completion date |
|---|---|---|---|---|---|---|---|
| NapaSinti trial (56) (ChiCTR2100052118) | China | Prospective, nonrandomized, open-label, phase II | 63 | Sintilimab + nab-paclitaxel | ORR | The target ORR for success is set at 15% or higher | March 2026 |
| Brightline-2 (34) (NCT05512377) | Multiple countries | Phase IIa/IIb | 155 | BI 907828 (45 mg) orally on day 1 of a 3-week cycle | ORR | Efficacy of brigimadlin in stabilizing p53 and promoting apoptosis or cell cycle arrest in TP53 wild-type tumor cells | March 25, 2027 |
| RAGNAR (NCT04083976) | Global | Phase II, open-label | 31 | FGFR1–4 inhibitor etafitinib in individuals with FGFR mutations (adults and children) | ORR | Temporary assessment: ORR: 41.9%, mDOR: not disclosed, DCR: not disclosed, mPFS: not disclosed, mOS: not disclosed | December 2027 |
| TRITICC (NCT04059562) | Germany | Phase II, open-label | 28 | Trifluridine/tipiracil + irinotecan | PFS | Not reported | October 2023 |
| ABC-06 (NCT01926236) | Global | Phase III, open-label | 81 | mFOLFOX vs. active symptom control | OS | ORR: 5%, DCR: 33%, mPFS: 4 months, mOS: 6.2 vs. 5.3 months | January 2018 |
| NIFTY (NCT03524508) | South Korea | Phase IIb open-label, 1:1 randomized | 88 | Nal-IRI + 5-FU/LV vs. 5-FU/LV | PFS | ORR: 14.8% vs. 5.8% (all complete response), DCR: 34.9% vs. 64.8%, mPFS: 7.1 vs. 1.4 months, mOS: 8.6 vs. 5.5 months | September 2020 |
| NALIRICC (NCT03043547) | Germany | Phase II, open-label, 1:1 randomized | 49 | 5-FU/LV vs. nal-IRI + 5-FU/LV | PFS | ORR: 5%, DCR: 33%, mPFS: 4 months, mOS: 6.2 vs. 5.3 months | December 2021 |
| NAPOLI-2 (NCT04005339) | USA | Phase II, open-label | 25 | 5-FU/LV + nal-IRI | PFS | ORR: 4%, DCR: 33%, mPFS: 4.0 months, mOS: 8.6 months | November 2023 |
| LEAP-005 (NCT03797326) | Global | Phase II, open-label, non-randomized | 31 | Pembrolizumab + lenvatinib in different tumors, including BTC | ORR | ORR: 4%, DCR: 33%, mPFS: 4.0 months, mOS: 5.3 vs. 6.2 months | December 2023 |
| CA209-538 (NCT02923934) | Australia | Phase II, open-label | 39 | Nivolumab with ipilimumab in several cancers, including BTC | DCR | mPFS: 2.9 months, mOS: 5.7 months, DCR: 44%, ORR: 23% (all partial response) | April 2020 |
| MCC-17651 (NCT02115542) | USA | Phase II open-label | 43 | Regorafenib | ORR | ORR: 11% (all partial response), DCR: 56%, mPFS: 15.6% of a year (or approximately 3.9 months), mOS: 31.8 weeks or approximately 7.95 months | December 2018 |
| IMMUNOBIL-D18-1 PRODIGE 57 (NCT03704480) | France | Phase-II, open-label, 1:1 randomized | 106 | Arm A: durvalumab + tremelimumab | OS | Arm B terminated early due to toxicity | October 2022 |
| Arm B: weekly paclitaxel | Arm A: ORR: 9.7% (7.8% partial response), DCR: 40.8%, mPFS: 2.5 months, mOS: 8 weeks, 6-month mOS rate: 59.2% | ||||||
| ClarIDHy (NCT02989857) | Global | Phase III, double-blind, 2:1 randomized | 124 | Ivosidenib rather than placebo in individuals with IDH1 mutations | PFS | ORR: 2% vs. 0% for partial response, DCR: 51 vs. 28%, mPFS: 2.7 vs. 1.4 months, mOS: 7.5 vs. 10.3 months, crossbreeding adjusted mOS: 5.1 vs. 10.3 months | January 2019 |
| NCT03212274 | USA | Phase II, open-label | 145 | Olaparib in patients with IDH1 or IDH2 mutations, such as CCC | ORR | Not reported | July 2023 |
| FIGHT-202 (NCT02924376) | Global | Phase II, open-label, nonrandomized | 107 | Pemigatinib (an FGFR1–3 inhibitor) for patients negative for FGF/FGFR mutations but not for those with FGFR2 fusion or other FGF/FGFR mutations | ORR | FGFR2 rearrangements or fusions: mOS: 21.1 months, mPFS: 6.9 months, ORR: 35.5%, DCR: 82.2%, mDOR: 7.5 months. Additional FGF/FGFR changes: mOS: 6.7%, mPFS: 2.1 months, DCR: 40.0%, ORR: 0%. Negative for modification of FGF/FGFR: mOS: 4 months, mPFS: 1.7 months, ORR: 0%, DCR: 22.2% | February 2022 |
| NCT02150967 | Global | Phase-II | 108 | Infigratinib (FGFR1–4 inhibitor) for patients with FGFR2 change | ORR | ORR: 23.1%, second-line ORR: 34%, third-line ORR: 13.8%, DCR: not recorded, mPFS: 7.3 months, mDOR: 5.0 months | July 2022 |
| HERIZON-BTC-01 (NCT04466891) | Global | Phase IIb, open-label | 100 | Bispecific antibody zanidatamab in HER2-positive patients with BTC | ORR | ORR: 41.3%, mDOR: 12.9 months, PFS: 6 months, mOS: immature | December 2022 |
| NCT04837508 | China | Phase II, open-label | 86 | MRG002 (ADC) in patients with HER2-positive BTC | ORR | Not reported | December 2022 |
| NCT03185988 | China | Phase II, open-label | 100 | Trastuzumab with chemotherapy for those with HER2-positive tumors, including BTC | ORR | Not reported | July 2017 |
| NCT04042831 | USA | Phase II, open-label | 100 | Olaparib for BTC with DDR mutation | ORR | Not reported | September 2022 |
| KEYNOTE-158 (NCT02628067) | Global | Phase II, open-label | 104 | Pembrolizumab | ORR | ORR: 5.8%, DCR: not reported in this cohort (not specified), mPFS: 2.0 months, mOS: 7.4 months, mDOR: not reached | June 2026 |
| CodeBreak100 | Global | Phase I–II | 129 | Sotorasib for patients with solid tumors with KRAS G12C mutations | ORR | Insufficient sample size with (only 1 patient with BTC) | May 2027 |
5-FU, 5-fluorouracil; ADC, antibody–drug conjugate; BTC, biliary tract cancer; CCC, cholangiocellular carcinoma; DCR, disease control rate; DDR, DNA damage response; FGF, fibroblast growth factor; FGFR, fibroblast growth factor receptor; HER2, human epidermal growth factor receptor 2; LV, leucovorin (also known as folinic acid); mDOR, median duration of response; mFOLFOX, modified folinic acid (leucovorin); mOS, median overall survival; mPFS, median progression-free survival; nal-IRI, nanoliposomal irinotecan; ORR, objective response rate.
A phase II trial on the efficacy of anlotinib and sintilimab as second-line therapy for patients with advanced BTC was conducted, with the primary endpoint being the OS rate. The treatment for this study was administered to 20 consecutively enrolled patients. The median OS for the trial was 12.3 months (95% CI: 10.1–14.5), there were no grade 4 or 5 treatment-related adverse events (TRAEs), and only 4 patients (20%) had grade 3 TRAEs. Sintilimab and sintilimab demonstrated a manageable safety profile and promising antitumor efficacy (57). FOLFOX is currently the recommended second-line treatment for unselected patients without advanced BTC. For those with advanced BTC, the focus should be shifted to immunotherapy and targeted therapy.
Targeted therapy for BTCs
To be able to treat patients with advanced BTCs successfully and improve their outcomes, new therapeutic approaches must be developed. Novel research on BTC therapies has included a focus on tyrosine kinase receptor-related pathways, epigenetic silencing, and certain gene alterations (58). By identifying potential genetic alterations, deep sequencing studies have shed light on the complicated molecular biology involved in BTCs (59,60). There is a comparatively high prevalence of druggable genetic modification in patients with advanced BTC, and they have demonstrated encouraging responses to molecular-targeted treatments for gene abnormalities such as FGFR2 fusions and rearrangements, IDH1 mutations, and HER2 overexpression and amplification (61). A comprehensive analysis found that approximately 40% of patients with BTC harbor potentially actionable genetic alterations. Notably, the distribution of driver mutations varies among the different subtypes of cholangiocarcinoma (62). Targeted therapy is used to treat BTCs in most cases that are resistant to chemotherapy. BTCs have been linked to several carcinogenic pathways, especially in iCCAs, which show more aggressive changes than do eCCAs, and exhibit more effective mutations than do GBCs (63). Targeted therapies have significantly extended the array of possible treatment choices for individuals with refractory illness and prolonged survival in those with advanced BTCs. The NER may serve as a prognostic biomarker in BTs, aiding treatment stratification in immunotherapy and targeted therapy settings (13). Given the advancements in the diagnosis of genetic changes, the treatment landscape for patients with advanced BTCs remains dynamic (64).
Isocitrate dehydrogenase inhibitor
IDH is an enzyme that transforms isocitrate to α-ketoglutarate in the citric acid cycle. Mutant IDH1, along with IDH2, then converts α-ketoglutarate to the carcinogenic metabolite, 2-hydroxyglutarate (2-HG). The buildup of 2-HG causes the disruption of cellular metabolism. IDH1 mutations are far less rare than are IDH2 mutations (21,65,66), and a common mutation in patients with cholangiocarcinoma involves the IDH, the metabolic enzyme (67,68). About 20% of those with iCCA have IDH1 mutations. In the phase III ClarIDHy trial, the median PFS was 5.1 months (95% CI: 3.8–7.6) with ivosidenib and 1.6 months (95% CI: 1.4–1.8) with placebo, while the median OS was 10.3 months (95% CI: 7.8–12.4) and 7.5 months (95% CI: 6.2–8.3), respectively (69). IDH1, IDH2, and pan-IDH1/2 inhibitors are among the various IDH inhibitors that are being tested in clinical trials for the treatment of iCCA. Early-phase clinical trials have produced the first encouraging results. For instance, a phase I research applied a 500-mg daily dose of ivosidenib, an IDH1 inhibitor, and successfully assessed 77 individuals with CCA who had previously received treatment but had IDH1 mutations (67). The phase III ClarIDHy trial enrolled 185 patients with metastatic IDH1-mutated cholangiocarcinoma and compared ivosidenib to placebo as second- or third-line therapy, with PFS as the primary endpoint. Overall, 230 participants were evaluated between February 20, 2017 and January 31, 2019. At the cutoff date, 185 patients were randomized to receive either placebo (n=61) or ivosidenib (n=124). The median PFS of the ivosidenib group was 6.9 months (IQR, 2.8–10.9 months), representing a considerable improvement as compared with the placebo group [one-sided P<0.0001; median OS 2.7 months (95% CI: 1.6–4.2) vs. 1. 4 months (95% CI: 1.4–.6); HR =0.37, 95%: CI 0.25–0.54], Ascites occurred in 4 of 59 patients receiving placebo and 9 of 121 patients receiving ivosidenib (7%), with grade-3-or-beyond ascites being the most common grade in both treatment groups. Moreover, 36 of 121 (30%) patients receiving ivocidinib and 13 of 49 (22%) receiving placebo experienced adverse events (AEs). Overall, ivosidenib was well tolerated and provided prolonged PFS as compared to placebo. Overall, targeting IDH1 mutations in patients with advanced IDH1-mutant cholangiocarcinoma has shown therapeutic benefit.
Common AEs of ivosidenib include severe fatigue, diarrhea, and nausea, while serious AEs include diarrhea, bleeding, elevated blood bilirubin level, and hyponatremia (70). Currently, the NCCN guidelines suggest oral IDH1 inhibitor ivosidenib as a second-line therapy for advanced, unresectable, and metastatic BTCs with mutations (71). This recommendation was made after the US Food and Drug Administration (FDA) approved the drug based on the above-mentioned phase III randomized controlled ClarlDHy trial (70). Ivosidenib is currently being evaluated in an ongoing phase I study (NCT04088188) as a component of first-line combination therapy along with GC, which is expected to be finished in 2025. Early-phase trials are also being conducted on additional IDH inhibitors. LY3410738, an oral medication that is already thought to be more effective and perhaps more long-lasting than ivosidenib, is presently being studied in a phase I trial (NCT04521686) for certain BTC IDH variants (IDH2-R140, IDH1-R132, and IDH2-R172). Trials investigating the tolerability of IDH305, an investigational oral drug reserved only for malignancies containing IDH1-R132 mutations, are now underway (NCT02381886). According to preliminary evidence, the oral inhibitor of mutant IDH1, olinusidenib, has shown modest clinical activity, safety, and tolerability, with 23% of treated iCCA patients exhibiting stable disease (72,73). An oral dual IDH1 and IDH2 inhibitor, voracidenib, is also under development, and although its clinical activity in gliomas has been demonstrated in preliminary trials, its effect in BTC has not yet been established (24,74). FDA approved ivosidenib in August 2021 for previously treated patients with CCA and who had a confirmed IDH1 mutation according to an FDA-approved test and who were either locally progressed or metastatic. Moreover, ivosidenib and nivolumab were both tested in combination in a phase II trial for advanced solid tumors with IDH1 mutations (NCT04056910). Regarding IDH2 inhibition, a number of drugs are presently under investigation in early-phase trials, including AG-881 (NCT02481154), which is examining IDH1/2 inhibitors (75).
FGFR inhibitor
The four receptor tyrosine kinases (FGFR 1 to 4) that make up the family known as FGFRs are encoded by the same gene. This receptor exerts several biological functions through several signaling pathways, including the RAS/MAPK and PI3K.AKT pathways (76). This family of oncogenic changes (mutations, fusions, amplifications, or rearrangements) is observed in 7% of solid tumors and is linked to aberrant activation of FGFR, which causes angiogenesis, cell division, and immune escape (76,77). iCCA with FGFR2 fusion has been associated with female gender, young age, and the presence of a concurrent BAP1 mutation (59,78). Approximately 15% of individuals with iCCA have FGFR2 rearrangements or fusions. TAS-120, pemigatinib, infigratinib, and Debio 1347 are among the FGFR inhibitors being investigated in the treatment of BTCs (17,79-81). The FGFR inhibitor infigratinib was assessed in 71 patients with advanced BTCs in a phase II trial. With infigratinib, the ORR was 31%, the median time frame of response was 5.4 months, the median PFS was 6.8 months, the median OS was 12.5 (95% CI: 9.9–16.6) months, and the disease control rate (DCR) was 83.6% (80). Pemigatinib, a selective reversal inhibitor of FIGR 1–3 was approved for treating advanced BTCs after the phase II FIGHT 202 trial demonstrated its efficacy, with an ORR of 35.5% (95% CI: 26.5–45.4%) and with three patients who were unresponsive to previous treatment achieving CR. (82). The highly anticipated phase III Fight 302 (NCT03656536) randomized controlled trial comparing pemigatinib with GC as first-line therapy in advanced BTC is currently underway. Futibatinib is a unique, extra-potent oral drug that acts as an irreversible, covalent pan-inhibitor of FGFR 1–4. In the phase II FOENIX CCA2 trial, it yielded a 12-month survival rate of 73% and an ORR of 41.7% (83). The ongoing FOENIX CCA3 trial (NCT04093362) is currently investigating the efficacy and safety profile of futibatinib as compared to GC as first-line therapy. The first selective FGFR2 inhibitor, RLY-4008, was designed to reduce on-target toxicity and bypass the polyclonal FGFR2 resistance pathway (66). Evidence from the phase I/II ReFocus trial (NCT04526106), presented at the European Society for Medical Oncology (ESMO) Congress 2022, indicated that RLY-4008 has strong efficacy, with an ORR of 88% (95% CI: 63.6–98.5%) in the first 17 patients with FGFR-naïve BTC who received the recommended phase II treatment (66). Enrollment in additional cohorts consisting of patients with BTCs exposed to FGFR inhibitors and all solid tumors is also ongoing (66). Table 6 provides a summary of the completed and ongoing trials for FGFR inhibitors.
Table 6
| Clinical trial | Country | Phase and design | Enrolment/estimated enrolment, n | Interventions | Completion time/estimated completion time |
|---|---|---|---|---|---|
| FIGHT-302 (NCT03656536) | UK | Phase II: randomized, active-controlled, open-label, multicenter | 434 | Pemigatinib vs. cisplatin with gemcitabine | July 2028 |
| FOENIX-CCA3 (NCT04093362) | USA | Phase III study, open-label, randomized | 216 | Gemcitabine vs. cisplatin in futibatinib | June 2024 |
| ReFocus trial (NCT04526106) | USA | Phase I/II, FIH trial, open-label | 540 | RLY-4008 | October 2024 |
FGFR, fibroblast growth factor receptor; FIH, factor inhibiting hif (hypoxia-inducible factor).
HER2
The overexpression of HER2 (also known as ERBB2) occurs in 15% to 20% of BTC cases, especially in eCCA and GBC (84). The combination of the monoclonal antibodies pertuzumab, along with trastuzumab, has been extensively investigated. Although it has not received FDA approval, it is included in the NCCN guidelines as a treatment option for patients with malignancies that are positive for HER2 after first-line therapy (71). A large multibatch phase II clinical trial, MyPathway (NCT02091141), is assessing the efficacy of trastuzumab and pertuzumab in the treatment of patients with metastatic biliary tract tumors with HER2 overexpression and amplification that have not responded to prior therapies. Although the initial results were unsatisfactory, further updates with additional enrollment include an ORR of 23% (95% CI: 11–39%) (85,86). Trastuzumab deruxtecan (T-DXd) is being assessed in the prospective HERB 2 trial in Japan for the treatment of patients with HER2-mutant cholangiocarcinoma who are intolerant or unresponsive to gemcitabine as first-line therapy (87). T-DXd is an antibody drug combination composed of deruxtecan, a topoisomerase inhibitor, a cleavable linker, and the anti-HER2 antibody trastuzumab. The preliminary data presented at the 2022 ASCO Annual Meeting 202 2are promising, with a DCR of 81.8% (95% CI: 59.7–94.8%) and an ORR of 36.4% (95% CI: 19.6–56.1%). However, major side effects were noted, with 25% of patients developing interstitial lung disease, severe gastrointestinal toxicity, and myelosuppression, all of which require further investigation and observation (88). SGNTUC-019 (NCT04579380) was an open-label phase II trial aimed at assessing the safety and efficacy of trastuzumab and tucatinib in patients with advanced malignancies harboring HER2 mutations. Among the 30 included patients, the median follow-up period was 10.8 months, with the data cutoff date being January 30, 2023. Patients were treated with trastuzumab (8 mg/kg intravenously and then 6 mg/kg every 3 weeks) and tucatinib (300 mg orally twice a day) in a 21-day cycle. Among 76.7% of patients (90% CI: 60.6–88.5%), the DCR was 46.7% (90% CI: 30.8–63.0%), the PFS was 5.5 months (90% CI: 3.9–8.1), and the median duration of response was 6.0 months (90% CI: 5.5–6.9). The 12-month OS rate at the data cutoff point was 53.6% (90% CI: 36.8–67.8%), and 15 patients (50.0%) died. The most common treatment-emergent adverse events (TEAEs) were diarrhea (40.0%) and pyrexia (43.3%). Eighteen patients (60.0%) had grade ≥3 TEAEs, most of which included fatigue, cholangitis, and nausea (10.0%), all of which were unrelated to the drug. One patient’s treatment regimen was discontinued due to a TEAE, but the TEAE did not result in the patient’s death. The results showed that previously treated HER2-positive patients with BTC responded to tutunitinib and trastuzumab, demonstrating the clinically significant anticancer activity of this regimen (89). There remain numerous challenges related to HER2-targeted therapy for BTC. It is interesting to note that, in contrast to FGFR and IDH, HER2 amplification is not relatively uncommon in patients with BTCs. Consequently, only a small number of these patients are suitable for clinical study trials, and more research is needed to gain insights into HER2 therapies in clinical settings (90).
B-raf kinase
The B-RAF protein encoded by BRAF is a member of the RAF family of serine threonine protein kinases. In the RAF, MEK, and ERK pathways, this protein functions as a primary enzyme, with tumorigenesis being associated with the dysregulation of these pathways.
Among patients with iCCA, 4% have BRAF mutations, representing the most common mutation type (91-93). Meanwhile, the BRAF V600E mutation is present in approximately 5% of patients with BTC. Combination therapy has considerably potential in the first-line treatment of patients with the BRAF V600E mutation. The Rare Oncology Agnostic Research (ROAR) trial (NCT02034110), a phase II, open-label, multicenter trial, evaluated the safety and efficacy of trametinib (an oral MEK inhibitor) and dabrafenib (an oral BRAF inhibitor) in the treatment of 43 patients with BTC and BRAF mutations who had progressed after chemotherapy (18). According to an independent observer evaluation, BTC-related cases had an ORR of 47%, a median PFS of 9 months, and a median OS of 14 months. Reported AEs included fatigue, flushing, nausea/vomiting, and pyrexia (18).
Vemurafenib is an oral selective RAF inhibitor for BRAF V600 mutations. In a multicenter basket study of 172 patients with BRAF V600-mutant solid tumors excluding melanoma, 9 of the patients had BTCs, and 99% of the patients had BRAF V600E mutations. An ORR of 33.3% was recorded among the patients with BTCs (94,95). In June 2022, the US FDA approved dabrafenib in combination with trametinib for the treatment of patients with advanced solid tumors and the BRAF V600E mutation (96). ABM 1310, a novel small oral molecule inhibitor of BRAF V600E, is presently being evaluated in phase I trials for its safety and potential antitumor efficacy. Concurrently, studies are being carried out for ABM 1310’s combination with the MEK inhibitor cobimetinib (NCT04190628) and monotherapy (NCT05501912). Drugs targeting non-BRAF V600E-mutant melanoma are also being evaluated, with the FDA-approved BRAF-MMEK inhibitor combination yielding promising results (97).
Neurotrophic tropomyosin receptor kinase (TRKs) inhibitors
The NTRK genes, NTRK1, NTRK2, and NTRK3, encode the TRKs, TRKA, TRKB, and TRKC, respectively. These tyrosine kinase receptors are critical to the proper development of the nervous system (98). NTRK gene rearrangements occur in about 3% of patients with iCCA (99). With an overall 75% response rate and prolonged remission, the TRK inhibitor larotrectinib has yielded encouraging results in TRK fusion-positive patients with advanced malignancies, such as BTCs (100). Neurotrophins normally activate TRK receptors and, through ERK signaling, promote cellular growth and multiplication. Meanwhile, fusion rearrangements create hybrid genes by placing constitutively active genes adjacent to NTRK sequences. This results in uncontrollably activated ERKs and ligand-independent TRK receptor signaling (101). Through this key mechanism, TRK-binding proteins are recognized as oncogenic drivers in many tumors. They are more common in rare cancers such as secretory breast carcinoma, where they appear in over 90% of cases, as compared to in fewer than 1% cases of common cancers such as lung and colorectal cancer. The incidence of TRK-binding proteins in bile duct carcinoma is estimated to be around 0.25% (102). A phase I–II trial evaluated larotrectinib treatment in 159 patients with NTRK fusion-positive solid tumors who had previously received conventional therapy. The ORR was 79%, the CR rate was 16%, the median PFS was 28.3 months, and the OS was 44.4 months. One of the two patients with BTC exhibited a limited response, while the other’s disease worsened over time (103). Larotrectinib is an encouraging alternative treatment for patients with BTCs and the NTRK fusion, although NTRK fusion has been documented in only 0.2–0.7% of patients with this disease (102,104).
Immunotherapy
Immunotherapy mainly consists of ICIs that target cytotoxic T-lymphocyte antigens, PD-1, and PD-L1, as well as cancer vaccines and CTLA-4 cell transplants. Immunotherapy has been applied in treating various solid tumors, such as breast cancer, colon cancer, and non-small cell lung cancer. Immunotherapy is currently gaining popularity as a potent therapeutic approach for cancer and is a crucial component of systemic treatment for BTCs and other cancers. In immunotherapy trials, pembrolizumab, nivolumab, and durvalumab monotherapies have shown encouraging outcomes for patients with advanced BTC. Thus, combining immunotherapy with other treatments could be a useful strategy for increasing antitumor efficacy (90). ICIs, as one of the most common forms of immunotherapy, have been well researched in GBC, iCCA, and eCCA. In tumor immunotherapy, endogenous anticancer activity is primarily enhanced by monoclonal antibodies. The majority of the targets of these monoclonal antibodies are immunological checkpoint regulators, also known as ICIs (105). The most studied ICI targets are CTLA-4 and PD-1, as these are the most common T-cell immune checkpoints. In addition, ICIs that directly target other immunological checkpoints, such as LAG-3, TIM-3, TIGIT, and B7-HH3 are being developed (106). ICIs have demonstrated favorable treatment responses across a variety of tumor types (107-109). The US FDA has approved pembrolizumab and nivolumab for the treatment of advanced-stage cancers (110,111). Moreover, cytokine treatment and adoptive cell therapy (ACT) primarily target iCCA and have a moderate impact, but for GBC and eCCA, the efficacy is less conclusive (112). The 2022 phase III TOPAZ 1 trial assessed the efficacy of durvalumab immunotherapy in addition to standard GC chemotherapy in treating patients with recurrent BTC. The median OS increased from 11.5 to 12.8 months, and the ORR increased from 18.7% to 26.7% (39). A single-arm multicenter phase II trial assessed the efficacy of the PD-1 inhibitor nivolumab in the treatment of 54 patients with advanced BTC who had not responded to one or three lines of therapy. The DCR was 59%, and the investigator-assessed response rate was 22%. Patients who responded to therapy did not reach the median duration of treatment. The OS was 14.2 months, and the PFS was 3.7 months. Notably, the long PFS was associated with PD-L1 expression in tumor cells (HR =0.23, 95% CI: 0.10–0.51; P<0.001) (113).
A case report was published that indicated the potential of immunotherapy conversion for tumors that were initially considered incurable. The report described a patient with iCCA who underwent successful surgery following six cycles of ICI plus GEMOX, which decreased tumor size and enabled surgical resection (114). PD-L1 is the most common therapeutic target for cancer immunotherapy. Solid tumors with MSI-H status respond well to immunotherapy consisting of PD-L1 inhibitors (115,116). Pembrolizumab, which binds to and inactivates PD-1, has been approved for the treatment of various solid cancers with microsatellite instability. The safety and efficacy of pembrolizumab in the treatment of advanced BTCs were evaluated in the KEYNOTE 158 (phase II) and KEYNOTE 028 (phase Ib) trials. In 6% to 13% of patients with advanced BTC, monoclonal antibodies, independent of PD-L1 expression, demonstrated moderate toxicity and long-lasting anticancer activity (117). Several phase II trials have investigated another PD-L1 blocker, nivolumab, reporting encouraging results and a reasonable safety profile, both alone and with GC (118). In a phase II trial that enrolled 46 patients with advanced BTC who received at least first-line chemotherapy, nivolumab treatment yielded an ORR of 11% overall, a PFS of 3.7 months, and an OS of 14.2 months (113). Clinical trials are currently being conducted on the anti-CTLA-4 medication ipilimumab to evaluate its efficacy in treating various solid cancers. The CA209-538 (phase II) clinical trial reported that a combination of nivolumab and ipilimumab yielded favorable results in patients with advanced BTC. Compared to single-agent anti-PD-1 therapy, this combination was associated with higher response rates, with a 23% ORR among patients with GBC or iCCA; however, patients with eCCA did not respond to this treatment. The OS was 5.7 months (95% CI: 2.7–11.9). However, further trials are needed to determine if this combination immunotherapy regimen can provide superior therapeutic effect for patients with advanced BTC as compared to single-agent anti-PD-1 treatment (119).
The efficacy of immunotherapy as a first-line treatment has been evaluated in chemotherapy-naïve patients with BTC. Patients with a histologically confirmed diagnosis of advanced or unresectable BTCs were enrolled in the phase III KEYNOTE-966 trial (NCT04003636), and the OS of patients treated with GC plus pembrolizumab was compared with that in patients treated with GC alone. Although definitive findings are still expected, the preliminary results are encouraging, as GC plus pembrolizumab yielded a statistically significant increase in survival as compared to chemotherapy alone. The TOPAZ 1 trial (NCT03875235) was a double blind, placebo-controlled study that examined the effects of GC and durvalumab, an anti–PD L1 agent, in chemotherapy-naïve patients with advanced BTC. Patients who received durvalumab also showed significant improvements in PFS (HR =0.75, 95% CI: 0.64–0.89) and OS (HR =0.80, 95% CI: 0.66–0.97). Moreover, those who received durvalumab had an ORR of 26.7%, while those who received placebo had an ORR of 18.7% (39). Based on the results of the TOPAZ 1 trial, the US FDA approved durvalumab for the treatment of locally advanced or metastatic BTC in older patients in combination with GC chemotherapy. Additionally, the NCCN guidelines now recommend this regimen as first-line therapy for patients with metastatic, locally advanced or unresectable cancers.
Immunotherapy has emerged as a promising strategy in the treatment of BTCs, particularly with the introduction of ICIs such as durvalumab and pembrolizumab. Phase III trials such as TOPAZ-1 and KEYNOTE-966 have demonstrated improved survival outcomes when these agents were combined with chemotherapy. Despite this progress, the lack of reliable predictive biomarkers remains a substantial challenge, highlighting the need for further research into personalized treatment strategies (120).
ICI monotherapy
Novel approaches to therapy are being developed. For instance, the ICIs durvalumab or pembrolizumab have been added to GC as a first-line systemic treatment, providing prolonged survival as compared to GC alone (121).
In one clinical trial (NCT01876511), pembrolizumab treatment resulted in satisfactory results for 86 patients with dMMR or MSI-H (including those with cholangiocarcinoma) (116). In one phase Ib clinical trial, 228 patients with high-grade cholangiocarcinoma and 4 patients with high-grade GBC, all of whom were PD-L1-positive—received pembrolizumab monotherapy. Three of the patients with cholangiocarcinoma and one with GBC achieved stable disease. Grade 3 toxicity was observed in 17% of cases, but no grade 4 events occurred. Moreover, the ORR was 17%, the median PFS was 1.8 months, and the median OS was 6.2 months. Of note, this trial confirmed that pembrolizumab exerts anticancer activity, is well tolerated, and provides satisfactory safety and efficacy (117). In a phase II study, 54 patients with advanced refractory BTC who had received at least one line of systemic therapy but no more than three lines were treated with nivolumab. More than half of patients with well-controlled disease had a median PFS of 3.68 months and a median OS of 14.22 months (113).
In addition to immune-specific ICIs, immune-simulating ICIs have also garnered considerable attention. An encouraging example is ABL501, which has stronger antibody efficacy than does anti-LAG-3 and anti-PD-L1 combination therapy because it can inhibit LAG-3 and PD-L1 simultaneously. ABL501 is currently undergoing first-in-human trials (NCT05101109) and has emerged as one of the viable options for cancer immunotherapy (122). Another emerging agent is M7824, a unique bifunctional fusion protein. M7824 is a monoclonal antibody against PD-L1 fused to the extracellular domain of human TGF-β receptor II. Due to its unique structure, M7824 can inhibit TGF-β and PD-L1 simultaneously (123). One study examined M7824’s response in Asian patients with cholangiocarcinoma. It yielded an ORR of 23%, but 63% of the patients experienced TRAEs (124). Sex-based differences in immune-related adverse events (irAEs) may influence BTC management, particularly as ICIs are increasingly used in the treatment of advanced BTC. Understanding sex-specific toxicity profiles could help tailor immunotherapy strategies for patients with BTC receiving ICIs (125). However, more research is needed to evaluate the general safety and efficacy of this strategy in immunotherapies.
Dual-ICI combination therapy
Given the limited efficacy of ICI monotherapy, combination immunotherapy is increasingly being studied in its ability to improve therapeutic response and elicit an immune response. Two ICIs combined in combination therapy have shown encouraging results in various solid cancers. In one phase II trial, 39 patients with advanced BTC were treated a combination of nivolumab and ipilimumab. The trial reported a 44% DCR and a 23% ORR, demonstrating the considerable benefit of dual combination ICI therapy over monotherapy (119). In an advanced-stage phase I trial, the combination of durvalumab and tremelimumab was examined in 65 patients with BTC (126), yielding an ORR of 10.8%, a median OS of 10.1 months, and a median PFS of 1.6 months. However, 23.1% of patients experienced TRAEs of grade 3 or above. Meanwhile, in a study of Japanese patients with hepatocellular carcinoma (HCC) and BTC, durvalumab-tremelimumab combination therapy provided the least encouraging response rates and survival results (127). Because these studies involved small sample sizes, care should be taken in interpreting these findings. Consequently, even while combination immunotherapies appear promising, especially those involving dual ICIs, further studies including larger and more varied patient groups are necessary to determine the actual efficacy and safety of these strategies in the management of BTCs.
Chemotherapy combined with ICIs
According to one trial (NCT03046862), which divided 124 patients with advanced-stage BTC into three treatment groups (GC → GC + durvalumab, GC + durvalumab, and GC + durvalumab + tremelimumab), the combination of GC with ICIs may provide therapeutic benefit (128). Oh et al. (129) examined the combination of immunotherapy and GC in treating BTC in a phase III TOPAZ 1 trial. They further investigated the use of PD-L1 inhibitors in combination with chemotherapy for the treatment of advanced CCA. In this trial, 685 participants were randomized to undergo durvalumab plus GC or placebo plus GC. The durvalumab-plus-GC group had a median OS of 12.8 months and a median PFS of 7.2 months, while in the GC-placebo group, the median OS and PFS were 11.5 and 5.7 months, respectively. Interestingly, although TRAEs were found in 64.9% patients in the control group (placebo + GC) and 62.7% patients in the treatment group (durvalumab + GC). The combination of chemotherapy and ICIs for advanced BTCs did not greatly improve survival, while other combinations—such as ICIs with targeted therapies or dual immunotherapy—have shown more promising outcomes. This suggests that exploring alternative combination strategies beyond chemotherapy may offer a more effective approach to improving treatment efficacy in advanced BTCs (129). Furthermore, numerous studies have evaluated the safety and therapeutic efficacy of GC in conjunction with nivolumab (118,130,131), capecitabine and oxaliplatin combined with pembrolizumab (CAPOX), and paclitaxel combined with tremelimumab and durvalumab for treating BTCs (132). These trials have yielded consistently positive outcomes, with the exception of one (NCT03704480). Taken together, these studies indicate that ICIs combined with chemotherapy may be a valuable approach for treating individuals with BTC, providing both a respectable safety profile and improved efficacy.
ICIs combined with targeted therapy
It has been shown that the combination of immunotherapy and molecular targeted therapy is highly effective. Immunotherapy’s ability to reverse immunosuppression can prolong the subsidence effect that targeted molecular therapy induces, increasing targeted therapy’s overall efficacy.
In one trial (NCT02443324), the efficacy of pembrolizumab in combination with ramucirumab (a VEGFR 2 monoclonal antibody) was evaluated in 26 patients with locally advanced, unresectable or metastatic BTC. Only 1 patient experienced a limited response, and the median OS was 6.4 months, a very poor outcome (133). Another trial (NCT03201458) compared atezolizumab monotherapy with the MEK inhibitor cobimetinib in patients with BTC who had not responded to first- or second-line therapy (134). Both therapy groups had very low ORRs; however, the combination group had a slightly improved median PFS. In a similar setting, a single-arm study investigated the safety and efficacy of pembrolizumab combined with lenvatinib in the management of patients with BTCs who had not responded to prior systemic therapy. In this trial, 25% of patients responded to treatment, the DCR was 78.1%, and clinical benefit rate was 40.5% (135,136). The clinical trials examining the immunotherapy regimens for BTC are summarized in Table 7.
Table 7
| Clinical trial number | Phase | Treatment line | Type of BTC | Estimated enrollment, n | Allocations | Study arms | Targeted pathways | Primary outcome | Status |
|---|---|---|---|---|---|---|---|---|---|
| Monotherapy | |||||||||
| NCT04157985 | III | First line | CCA | 578 | Randomized | Pembrolizumab or nivolumab or atezolizumab or ipilimumab or cemiplimab | PD-L1/PD-1 | PFS, time to next treatment | Recruiting |
| NCT02054806 | Ib | Second line or beyond | GBC, CCA | 477 | N/A | Pembrolizumab | PD-1 | Overall best response | Completed |
| NCT02829918 | II | Second line or beyond | GBC, CCA | 54 | N/A | Nivolumab | PD-1 | ORR following 4 treatment cycles | Active but not recruiting |
| NCT03999658 | II | Second line or beyond | GBC, CAA | 220 | Nonrandomized | STI 3031 | PD-L1 | ORR | Not yet recruiting |
| NCT02628067 | II | Second line or beyond | GBC, CAA | 1,609 | Nonrandomized | Pembrolizumab | PD-1 | ORR | Recruiting |
| NCT03849469 | I | Second line or beyond | ICC | 242 | Nonrandomized | XmAb 22841 + pembrolizumab | PD-1, CTLA-4 and LAG-3 | Tolerability, safety | Active but not recruiting |
| ICI + chemotherapy | |||||||||
| NCT04066491 | II/III | First line | CCA, GBC | 309 | Randomized | M7824 + GC, L1 and TGF-β plus placebo | PD-L1 | Safety, OS | Active but not recruiting |
| NCT03875235 | III | First line | CCA, GBC | 810 | Randomized | Durvalumab + GC, placebo + GC | PD-L1 | OS | Active but not recruiting |
| NCT04003636 | III | First line | CCA, GBC | 1,040 | Randomized | Pembrolizumab + GC, placebo + GC | PD-1 | OS | Active but not recruiting |
| NCT04027764 | II | First line | CCA, GBC | 30 | N/A | Toripalimab + S-1 + albumin paclitaxel | PD-1 | ORR | Recruiting |
| NCT03478488 | III | First line | CCA, GBC | 480 | Randomized | GEMOX + KN035 | PD-L1 | OS | Recruiting |
| NCT03704480 | II | Second line | CCA, GBC | 102 | Randomized | Durvalumab plus tremelimumab plus paclitaxel, durvalumab CTLA-4 | PD-L1 | PFS | Recruiting |
| NCT04295317 | II | N/A | ICC | 65 | N/A | SHR-1210, capecitabine | PD-1 | RFS | Recruiting |
| ICI + targeted therapy | |||||||||
| NCT04361331 | II | First-line | ICC | 60 | Randomized | Toripalimab + lenvatinib, lenvatinib + GEMOX | PD-1, TKIs | ORR | Active but not recruiting |
| NCT03895970 | IIb | Second line | CCA, GBC | 50 | N/A | Pembrolizumab + lenvatinib | TKIs, PD-1, | PFS, ORR, DCR | Recruiting |
| NCT02443324 | I | Second or later line | CCA, GBC | 298 | N/A | Pembrolizumab + ramucirumab | VEGFR-2, PD-1 | Tolerability, safety | Completed |
| NCT03475953 | I/II | Second or later line | CCA, GBC | 482 | Nonrandomized | Avelumab + regorafenib | TKIs, PD-L1 | ORR, RP2D | Recruiting |
| NCT04298008 | II | Third line | CCA, GBC | 26 | N/A | Durvalumab + AZD6738 | PD-L1, ATR/ATM | DCR | Recruiting |
| ICI + chemotherapy + targeted therapy | |||||||||
| NCT04300959 | II | First line | CCA, GBC | 80 | Randomized | Sintilimab + anlotinib + GC | PD-1, c-KIT kinase, PDGFR, FGFR, and VEGFR | 12-month OS rate | Recruiting |
| NCT05342194 | III | First line | ICC | 480 | Randomized | Toripalimab + lenvatinib + GEMOX/GC, placebo + GEMOX/GC | TKIs, PD-1 | OS | Not yet recruiting |
| NCT03951597 | II | First line | ICC | 30 | N/A | JS001 + lenvatinib + GEMOX | TKIs, PD-1 | ORR | Active but not recruiting |
| NCT05211323 | II | First line | ICC | 88 | Randomized | Atezolizumab + bevacizumab + GC | VEGFR, PD-1 | PFS | Recruiting |
| NCT04506281 | II | NAT | ICC | 128 | Randomized | Toripalimab + lenvatinib + GEMOX, capecitabine | TKIs, PD-1 | Event-free survival | Recruiting |
In the future, immunotherapy as a cornerstone therapy may be used in conjunction with targeted therapy or chemotherapy to treat BTCs. ATM, ataxia telangiectasia mutated; ATR, ataxia telangiectasia and rad3 related; BTC, biliary tract cancer; c-KIT, tyrosine-protein kinase KIT (also known as CD117); CCA, cholangiocarcinoma; CTLA-4, cytotoxic T lymphocyte-associated antigen 4; DCR, disease control rate; FGFR, fibroblast growth factor receptor; GBC, gallbladder cancer; GC, gemcitabine and cisplatin; GEMOX, gemcitabine and oxaliplatin; ICC, intrahepatic cholangiocarcinoma; ICI, immune checkpoint inhibitor; L1, L1 cell adhesion molecule (L1CAM); LAG-3, lymphocyte activation gene 3; N/A, not available; NAT, neoadjuvant therapy; ORR, objective response rate; OS, overall survival; PD-1, programmed cell death protein 1; PD-L1, programmed death ligand 1; PDGFR, platelet-derived growth factor receptor; PFS, progression-free survival; RP2D, recommended phase II dose; RFS, recurrence-free survival; TGF-β, transforming growth factor-beta; TKI, tyrosine kinase inhibitor; VEGFR, vascular endothelial growth factor receptor.
ACT
The term ACT describes the intravenous administration of immune cells that have been altered in the tumor or peripheral blood to patients with cancer in order to mediate an anticancer effect (137). Autologous tumor-infiltrating lymphocyte (TIL) infusion, chimeric antigen receptor T-cell (CAR-T) therapy aimed against particular surface tumor antigens, and genetically modified T-cell receptor (TCR) T-cell therapy are examples of such treatments (138). In 2006, a case report proposed the therapeutic efficacy of ACT in BTCs. A patient with iCCA who had lymph node involvement had extensive resection and was treated with adjuvant CD3-activated T cells with tumor lysate- or peptide-pulsed dendritic cells (DCs). He lived for more than 3.5 years after therapy (139).
Chimeric antigen receptor T cells and genetically modified TCRs
CAR-T cells are T cells produced from patients that have been genetically modified with exogenous receptors that identify tumor-expressing antigens. Technologies such as viral vector-mediated gene therapy and CRISPR/Cas9-mediated genome editing enable the insertion of recombinant DNA that codes CAR and the elimination of TCR gene expression. The patient is then administered CAR-T cells to destroy cancerous cells. Genetically engineered TCR treatments modify T-cell specificity by expressing particular TCR α and β chains, which facilitate antigen recognition. Tumor-specific TCR α and β chains are identified, extracted, and cloned into transduction vectors. T cells are then transduced to produce tumor-antigen-specific lymphocytes (140).
A number of phase I and II clinical studies of CAR-T treatment for advanced BTCs have now been completed (112). In a phase I clinical study of CAR-T-cell therapy targeting HER2 in nine patients with advanced BT, one had a partial response (141). As of this writing, three patients are in good physical health, indicating that CAR-T therapy may be a viable strategy for increasing the longevity of patients with BTC and expressing HER2. Another clinical study evaluated CAR-T treatment for patients with advanced BTC and overexpression of epidermal growth factor receptor (EGFR). Ten patients experienced disease control, and one patient was in total remission (142). Several countries have conducted clinical trials on CAR-T and TCR therapy (Table 8).
Table 8
| Origin country | Number of trials | Initiated | Currently active | Inactive |
|---|---|---|---|---|
| China | 470 | 433 | 273 | 197 |
| USA | 418 | 408 | 280 | 138 |
| UK | 43 | 43 | 26 | 17 |
| Germany | 15 | 15 | 11 | 4 |
| Belgium | 10 | 10 | 7 | 3 |
| France | 8 | 8 | 4 | 4 |
| Japan | 7 | 7 | 4 | 3 |
| Singapore | 9 | 7 | 3 | 6 |
| Australia | 3 | 3 | 2 | 1 |
| Italy | 4 | 4 | 4 | 0 |
| Netherlands | 3 | 3 | 2 | 1 |
| Spain | 3 | 3 | 3 | 0 |
| Russia | 2 | 2 | 2 | 0 |
| Israel | 2 | 2 | 2 | 0 |
| Sweden | 2 | 2 | 1 | 1 |
| Switzerland | 2 | 2 | 0 | 2 |
| Canada | 2 | 1 | 1 | 1 |
| Czech Republic | 1 | 1 | 0 | 1 |
| Malaysia | 1 | 1 | 1 | 0 |
| Norway | 1 | 1 | 0 | 1 |
| Turkey | 1 | 1 | 1 | 0 |
| Total | 1,007 | 957 | 627 | 380 |
This table was adapted from an open access article (143) under the terms of the Creative Commons CC BY license.
TILs
In TIL therapy, specific effector T cells are extracted from tumor tissue via TILs, cultivated in vitro, and subsequently administered to patients. Research has indicated that TILs may benefit patients with distant organ metastasis or locally progressed BTC (112). In a clinical experiment, TILs significantly inhibited BTC in a patient with an ERBB2-interacting protein mutation (144). Additionally, in 2012, a clinical trial (UMIN000005820) involving 62 BTC cases reported that the PFS of 36 patients with iCCA treated with surgery, DC vaccination, and TILs was 18.3 months, which was significantly longer than the 7.7 months of PFS observed in the 26 patients treated with surgery alone (P<0.05) (145).
Cytokine-induced killer (CIK) cell therapy
For CIK cell therapy, peripheral blood lymphocytes from patients are cultured in vitro and subsequently reinfused into the body to produce antitumor effects. In a clinical trial, 2 out of 85 patients achieved complete response (CR), 14 had partial response, and 54 had good curative outcomes when CIK cell therapy and DC vaccination were administered to patients with BTC (146).
Radiotherapy
Radiotherapy may be recommended for certain patients with BTC. However, according to the ESMO guidelines, there is little clinical evidence to support its efficacy. Radiotherapy can be used to ameliorate a number of the signs and symptoms of stomach cancer via the shrinking of tumors that block the bile ducts, blood vessels, or nerves (2). A retrospective analysis of patients with inoperable iCCA who underwent final radiation therapy revealed that the long-term survival rates appear to be comparable to resection at a biologically effective dosage (BED) of >80.5 Gy (147). Manterola et al. (148) conducted a review of the literature on radiotherapy alone or in combination with chemoradiotherapy (CRT) and fluoropyrimidines in an adjuvant setting as a radiosensitizer for resected GBC. Although adjuvant CRT’s efficacy remains controversial, the SWOG080 trial examined gemcitabine combined with capecitabine (Gem-cape) plus concomitant computed tomography-guided chemoradiation, which produced an excellent 2-year OS rate of 65% (95% CI: 53–74%) for 79 patients, regardless of the condition of resection limits (149). Unfortunately, a consensus definitive conclusion could not be reached regarding the radiation mode, dosage, or appropriate administration time due to the lack of direct comparisons and the poor quality and scarcity of data. Given that CRT involves modest radiation doses, it can be speculated that it will benefit OS more than other adjuvant treatments that have less harmful consequences (149). In the ASCO guidelines (150), the role of CRT is not specified, but some research suggests that it should be restricted to patients with eCCAs who have undergone R1 resection or who have other high-risk factors (151).
Local treatment
Transarterial chemoembolization (TACE)
TACE involves the administration of chemotherapeutic drugs directly into the arteries that feed the tumor. TACE increases the concentration of chemotherapeutic agents in the tumor while decreasing the concentration in normal tissues. As a result, TACE can enhance therapeutic efficacy while minimizing systemic toxicity. In recent years, research on the efficacy of TACE in patients with unresectable BTC has produced encouraging findings (152,153). TACE is a popular, minimally invasive treatment for patients with HCC, particularly those with intermediate-stage illness and a strong liver functional reserve (154). Furthermore, TACE has been assessed as a downstaging therapy for lowering tumor burden in order to transform previously unresectable HCC to possibly resectable illness (155). TACE has two tumor-suppressive functions: embolizing the branches of arteries feeding HCC to cause ischemic tumor necrosis and releasing chemotherapeutic medicines in a regulated and sustained manner to eliminate residual cancer cells (156). TACE has recently emerged as an effective postoperative adjuvant therapy for patients with HCC. A prospective randomized controlled trial found that postoperative TACE substantially reduced tumor recurrence, resulting in a longer OS and disease-free survival (DFS) for patients with hepatitis B virus-related HCC with an intermediate or high risk of recurrence. Importantly, the therapy was shown to be both safe and well-tolerated (157). In the TACTICS trial (NCT01217034), a randomized, prospective trial comparing TACE plus sorafenib to TACE alone in patients with unresectable HCC, TACE plus sorafenib did not show a significant OS (36.2 months) benefit over TACE alone (30.8 months); however, there was clinically meaningful OS prolongation and significantly improved PFS (22.8 months). Thus, the TACE with sorafenib might be considered a therapeutic option in intermediate-stage HCC, especially in patients with substantial tumor burden (158). Another phase III, randomized controlled trial (NCT03905967) aimed to evaluate clinical results of lenvatinib coupled with TACE (LEN-TACE) compared to lenvatinib monotherapy in patients with advanced HCC. Of the 338 patients, 170 were randomly assigned to receive LEN-TACE and 168 to receive lenvatinib monotherapy. Following a median follow-up of 17.0 months, the LEN-TACE group had a substantially longer median OS compared to the lenvatinib-alone group (17.8 vs. 11.5 months; HR=0.45; P<0.001). The median PFS in the LEN-TACE group was 10.6 months compared to 6.4 months in the lenvatinib-alone group (HR =0.43; P<0.001), and the LEN-TACE group had a significantly higher ORR (54.1% vs. 25.0%; P<0.001). These findings suggest that the addition of TACE to lenvatinib enhances clinical outcomes and might be effective as a first-line therapy for individuals with advanced HCC (159).
Transarterial radioembolization (TARE)
TARE, also known as selective internal radiation treatment (SIRT), is used to treat primary tumors and liver metastases. The treatment is based on the fact that tumors have greater arterial vascularization than does nontumor liver tissue. When microspheres containing beta-emitting nuclides are administered via a microcatheter into the artery supplying the tumor-containing liver tissue, the tumor dosages are greater than those supplied to nontumor liver tissue (160,161). Furthermore, TARE can be used as a bridge therapy to slow hepatic tumor growth and guarantee that patients survive the waiting period for liver transplantation without acquiring contraindications to this procedure (162,163). Y90 is the most commonly applied radioactive element in radioembolization. The unstable Y90 radioisotope decays into the stable element zirconium-90 via logarithmic beta decay, with a half-life of 2.67 days (64.2 hours). In doing so, it produces a high-speed electron, a beta particle, which causes direct cytotoxic annihilation of the target tumor. The tissue penetration of Y90 varies from 2.5 to 11 mm, and a radiation dosage of up to 170 Gy may be delivered (164).
Hepatic artery infusion (HAI)
The combination of HAI and systemic chemotherapy has demonstrated clinical benefit in extending OS in patients with unresectable iCCA, and it has been used as a therapeutic option in a minority of patients. However, the lack of surgeons and oncologists with extensive expertise in using HAI chemotherapy limits its application (165). Poor hepatic function, extended systemic chemotherapy, substantial liver tumor burden, portal hypertension, portal vein thrombosis, and hepatic artery blockage are all absolute contraindications to HAI treatment (166). HAI chemotherapy was first designed to treat colorectal liver metastases (167,168). However, over the past two decades, additional data have been published regarding iCCA therapy (Table 9).
Table 9
| Study | Patients, n | Treatment regimen | PFS | OS |
|---|---|---|---|---|
| Jarnagin et al., 2009 (169) | Total: 34; iCCA: 26, HCC: 8 | 4-week cycle: HAI, FUDR (0.16 mg/kg × 20/pump flow rate) and DEXA 25 mg on day 1 for 14 days of each cycle | 7.4 months | 29.5 months |
| Kemeny et al., 2011 (170) | Total: 22; iCCA: 18, HCC: 4 | 4-week cycle: HAI, FUDR (0.16 mg/kg × 30/pump flow rate) and DEXA 25 mg on day 1 for 14 days of each cycle; SYS, bevacizumab 5 mg/kg every other week | 8.45 months | 31.1 months |
| Ghiringhelli et al., 2013 (171) | iCCA: 12 | Second-line treatment, 2-week cycle: HAI, GEM (1,000 mg/m2 given over 30 min) followed by OX (100 mg/m2 given over 2 h) | 9.2 months | 20.3 months |
| Konstantinidis et al., 2016 (172) | iCCA: 104; treated with HAI/SYS: 78; treated with SYS: 26 | 4-week cycle. HAI, FUDR (0.16 mg/kg × 20/pump flow rate) and DEXA 25 mg on day 1 for 14 days of each cycle; SYS, mostly GEM-based | HAI/SYS: 12 months; SYS: 7 months | HAI/SYS: 30.8 months; SYS: 18.4 months |
| Cercek et al., 2020 (173) | iCCA: 38 | 4-week cycle: HAI, FUDR (0.12 mg/kg × 30/pump flow rate) and DEXA 30 mg/pump on day 1 for 14 days of each cycle; SYS, GEM (800 mg/m2) with OX (85 mg/m2) on day 1 or 15, every 2 weeks | 11.8 months | 25.0 months |
This table was adapted from an open access article (168) under the terms of the Creative Commons CC BY license. DEXA, dexamethasone; FUDR, floxuridine; GEM, gemcitabine; HAI, hepatic arterial infusion; HCC, hepatocellular carcinoma; iCCA, intrahepatic cholangiocarcinoma; OS, overall survival; OX, oxaliplatin; PFS, progression-free survival; SYS, systemic.
There is little information on the use of localized treatment in unresectable iCCA in the current guidelines due to a paucity of prospective studies. The rigorous examination of these techniques in clinical trials is critical, and future research should examine tailoring chemotherapy and regimens to selected targets in mutant iCCA.
Thermal ablation
With the development of advanced clinical technology, minimally invasive thermal ablation of tumors has become common practice. Percutaneous radiofrequency ablation, microwave ablation, cryoablation, and irreversible electroporation are being increasingly applied in the treatment of solid neoplasms, from the ablation of tiny, unresectable tumors to experimental treatments (174). Thermal ablation involves the breakdown of tissue via severe hyperthermia (high tissue temperatures) or hypothermia (low tissue temperatures). The temperature change is limited to a specific area in and around the tumor. Ablation procedures using both heat and cold can be administered, and there is no one best therapy for all clinical presentations. Thus far, radiofrequency ablation has been the predominant energy type used for hyperthermic ablation, but microwave ablation may eventually take its place. Laser ablation allows for accurate thermal monitoring via magnetic resonance imaging (MRI), whereas high-intensity focused ultrasound ablation allows for external energy delivery as well as MRI compatibility. Cryoablation is used to treat many of the same malignancies and is more evident on CT and ultrasound than is hyperthermic ablation, although it may not be appropriate for other cancers due to a lack of coagulation. Despite several years of clinical practice, the majority of ablation systems are in their first or second generation of development. Thermal tumor ablation is likely to become increasingly common as energy delivery technology improves (175).
Future steps and discussion
The treatment landscape for BTCs has undergone significant transformations, yet challenges remain in improving patient survival rates. BTCs are aggressive malignancies, often diagnosed at advanced stages, which severely limit the effectiveness of traditional therapies. Surgical resection offers the best opportunity for a cure; however, only a small subset of patients are eligible for this approach due to late-stage diagnoses. As a result, the focus has shifted toward systemic therapies aimed at prolonging survival and improving quality of life for patients with metastatic or unresectable BTCs.
Chemotherapy, particularly the combination of GC has long been the cornerstone of treatment. The establishment of GC as the first-line therapy has significantly improved survival outcomes. However, resistance to therapy and the inherent toxicity associated with chemotherapy remain considerable hurdles. Advancements in the molecular understanding of BTCs have led to the identification of key genetic mutations and alterations, such as IDH1 mutations, FGFR2 fusions, and HER2 overexpression, that drive tumor progression. These insights have paved the way for targeted therapies that offer more personalized treatment options.
In addition, immunotherapy has emerged as a groundbreaking approach in BTC treatment, offering new hope for patients whose options were previously limited. The incorporation of ICIs such as pembrolizumab and durvalumab in combination with chemotherapy has yielded promising results in clinical trials, prolonging PFS and offering more durable responses. Although these combination therapies hold immense promise, challenges remain in identifying the specific patient populations who will benefit the most and in managing irAEs.
The evolution of second-line therapies, such as FOLFOX and nal-IRI, has expanded treatment options for patients who have failed first-line therapies. These therapies, when used in combination with other agents, have shown significant efficacy in extending survival, but patient selection remains key to achieving optimal outcomes. The ongoing exploration of combination therapies, involving cytotoxic agents, targeted therapies, and immunotherapy, represents a promising direction for improving outcomes among patients with BTC. Moreover, while substantial progress has been made in treating advanced BTC, the complexity of its biology necessitates continued research into novel therapeutic agents and the development of biomarkers that can predict treatment responses and guide clinical decision-making.
One of the most promising areas in the treatment of BTCs is the reconsideration of dosing strategies, particularly in the combination of cytotoxic chemotherapy with targeted therapies and immunotherapies. Recent studies have reevaluated the dosing of chemotherapy and radiotherapy in combination therapies to optimize efficacy and reduce toxicity, resulting in noninferior or even improved outcomes in various cancers, including BTCs. For example, the combination of gemcitabine with nab-paclitaxel has shown promising results, facilitating the adjustment of dosing schedules to minimize toxicity while maintaining therapeutic efficacy. Similarly, lower doses of cytotoxic agents combined with ICIs have demonstrated improved survival outcomes without the severe side effects traditionally associated with chemotherapy. The TOPAZ-1 and KEYNOTE-966 trials investigating the addition of durvalumab and pembrolizumab to GC chemotherapy, respectively, showed that immune checkpoint inhibition combined with chemotherapy could offer a durable response.
Radiotherapy, traditionally used in the palliative setting, has also undergone advances in its integration with systemic therapies. Stereotactic body radiation therapy combined with chemotherapy or immunotherapy has shown promising results, particularly in select patients with BTC and oligometastatic disease. Future clinical trials should continue to investigate the optimal timing, dosing, and sequencing of radiotherapy to maximize its synergy with novel therapies.
Looking ahead, the future of BTC therapy lies in personalized treatment regimens, in which therapy is tailored based on the individual’s tumor profile, genetic mutations, and molecular markers. Clinical trials will continue to refine these treatment approaches, with a focus on optimizing the dosing and sequencing of chemotherapy, targeted agents, and immunotherapies. The identification and validation of biomarkers such as MSI-H, dMMR, and IDH1 mutations will enable more precise therapy and improve clinical outcomes.
The continued development of targeted agents, particularly those focused on genetic alterations such as IDH1 mutations, FGFR2 fusions, and HER2 overexpression, will further personalize BTC treatment. Precision medicine, guided by comprehensive genomic profiling, will allow clinicians to identify better which patients are most likely to respond to certain therapies. Additionally, the expansion of immune-based therapies, such as bispecific antibodies and novel immune modulators, will further enhance the antitumor activity of immunotherapy in BTCs.
In conclusion, while significant progress has been made in the treatment of BTCs, the future holds even more promise, with advancements in combination therapies, personalized medicine, and immunotherapy driving improved outcomes. The integration of optimized dosing strategies for chemotherapy and radiotherapy in combination with targeted therapies and immunotherapies will be key to improving survival rates and minimizing side effects. However, challenges remain, and ongoing clinical trials and translational research will be essential in shaping the next generation of treatments for this challenging and often deadly disease.
Conclusions
BTCs remain among the most aggressive and challenging malignancies, with historically limited treatment options and poor survival outcomes. However, the therapeutic landscape is rapidly evolving. Advances in molecular profiling have enabled the identification of actionable mutations, driving the development of targeted therapies that offer personalized, mutation-specific treatment strategies. Meanwhile, the integration of immunotherapy, particularly ICIs, with traditional chemotherapy regimens has yielded meaningful survival benefits in several landmark trials. As precision oncology gains momentum, the future of BTC management lies in biomarker-driven approaches, rational combination therapies, and individualized care models. Continued research into resistance mechanisms, novel molecular targets, and predictive biomarkers is essential to optimizing outcomes. A multidisciplinary, patient-centered approach will be critical in translating these scientific advances into real-world clinical success.
Acknowledgments
The authors sincerely thank the referenced studies and consortiums for contributing open-access datasets for this analysis.
Footnote
Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://jgo.amegroups.com/article/view/10.21037/jgo-2025-83/rc
Peer Review File: Available at https://jgo.amegroups.com/article/view/10.21037/jgo-2025-83/prf
Funding: This study was supported by
Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://jgo.amegroups.com/article/view/10.21037/jgo-2025-83/coif). The authors have no conflicts of interest to declare.
Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.
Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.
References
- Sutherland M, Ahmed O, Zaidi A, et al. Current progress in systemic therapy for biliary tract cancers. J Hepatobiliary Pancreat Sci 2022;29:1094-107. [Crossref] [PubMed]
- Valle JW, Borbath I, Khan SA, et al. Biliary cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2016;27:v28-37. [Crossref] [PubMed]
- Raman SP, Fishman EK. Abnormalities of the distal common bile duct and ampulla: diagnostic approach and differential diagnosis using multiplanar reformations and 3D imaging. AJR Am J Roentgenol 2014;203:17-28. [Crossref] [PubMed]
- Banales JM, Marin JJG, Lamarca A, et al. Cholangiocarcinoma 2020: the next horizon in mechanisms and management. Nat Rev Gastroenterol Hepatol 2020;17:557-88. [Crossref] [PubMed]
- Global Burden of Disease Cancer Collaboration. Global, Regional, and National Cancer Incidence, Mortality, Years of Life Lost, Years Lived With Disability, and Disability-Adjusted Life-years for 32 Cancer Groups, 1990 to 2015: A Systematic Analysis for the Global Burden of Disease Study. JAMA Oncol 2017;3:524-48. [Crossref] [PubMed]
- Wang H, Sun P, Baria K. The world-wide incidence of biliary tract cancer (BTC). J Clin Oncol 2020;38:585.
- Bertuccio P, Malvezzi M, Carioli G, et al. Global trends in mortality from intrahepatic and extrahepatic cholangiocarcinoma. J Hepatol 2019;71:104-14. [Crossref] [PubMed]
- Forner A, Vidili G, Rengo M, et al. Clinical presentation, diagnosis and staging of cholangiocarcinoma. Liver Int 2019;39:98-107. [Crossref] [PubMed]
- Vogel A, Saborowski A. Current and Future Systemic Therapies in Biliary Tract Cancer. Visc Med 2021;37:32-8. [Crossref] [PubMed]
- Ilyas SI, Khan SA, Hallemeier CL, et al. Cholangiocarcinoma - evolving concepts and therapeutic strategies. Nat Rev Clin Oncol 2018;15:95-111. [Crossref] [PubMed]
- Lamarca A, Edeline J, Goyal L. How I treat biliary tract cancer. ESMO Open 2022;7:100378. [Crossref] [PubMed]
- Eckel F, Schmid RM. Chemotherapy in advanced biliary tract carcinoma: a pooled analysis of clinical trials. Br J Cancer 2007;96:896-902. [Crossref] [PubMed]
- Sahin TK, Ayasun R, Rizzo A, et al. Prognostic Value of Neutrophil-to-Eosinophil Ratio (NER) in Cancer: A Systematic Review and Meta-Analysis. Cancers (Basel) 2024;16:3689. [Crossref] [PubMed]
- Sahin TK, Rizzo A, Aksoy S, et al. Prognostic Significance of the Royal Marsden Hospital (RMH) Score in Patients with Cancer: A Systematic Review and Meta-Analysis. Cancers (Basel) 2024;16:1835. [Crossref] [PubMed]
- Rimassa L, Personeni N, Aghemo A, et al. The immune milieu of cholangiocarcinoma: From molecular pathogenesis to precision medicine. J Autoimmun 2019;100:17-26. [Crossref] [PubMed]
- Boscoe AN, Rolland C, Kelley RK. Frequency and prognostic significance of isocitrate dehydrogenase 1 mutations in cholangiocarcinoma: a systematic literature review. J Gastrointest Oncol 2019;10:751-65. [Crossref] [PubMed]
- 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;9:1064-79. [Crossref] [PubMed]
- 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;21:1234-43. [Crossref] [PubMed]
- Valle JW, Lamarca A, Goyal L, et al. New Horizons for Precision Medicine in Biliary Tract Cancers. Cancer Discov 2017;7:943-62. [Crossref] [PubMed]
- Harding JJ, Cleary JM, Quinn D, et al. Targeting HER2 (ERBB2) mutation-positive advanced biliary tract cancers with neratinib: results from the phase 2 SUMMIT ‘basket’trial. J Clin Oncol 2021;39:320.
- Javle M, Bekaii-Saab T, Jain A, et al. Biliary cancer: Utility of next-generation sequencing for clinical management. Cancer 2016;122:3838-47. [Crossref] [PubMed]
- Eso Y, Shimizu T, Takeda H, et al. Microsatellite instability and immune checkpoint inhibitors: toward precision medicine against gastrointestinal and hepatobiliary cancers. J Gastroenterol 2020;55:15-26. [Crossref] [PubMed]
- Lamarca A, Macarulla T. Facts and Hopes in the Systemic Therapy of Biliary Tract Carcinomas. Clin Cancer Res 2024;30:3688-96. [Crossref] [PubMed]
- Valle JW, Kelley RK, Nervi B, et al. Biliary tract cancer. Lancet 2021;397:428-44. [Crossref] [PubMed]
- Bridgewater J, Fletcher P, Palmer DH, et al. Long-Term Outcomes and Exploratory Analyses of the Randomized Phase III BILCAP Study. J Clin Oncol 2022;40:2048-57. [Crossref] [PubMed]
- Lamarca A, Edeline J, McNamara MG, et al. Current standards and future perspectives in adjuvant treatment for biliary tract cancers. Cancer Treat Rev 2020;84:101936. [Crossref] [PubMed]
- Komaya K, Ebata T, Shirai K, et al. Recurrence after resection with curative intent for distal cholangiocarcinoma. Br J Surg 2017;104:426-33. [Crossref] [PubMed]
- Edeline J, Benabdelghani M, Bertaut A, et al. Gemcitabine and Oxaliplatin Chemotherapy or Surveillance in Resected Biliary Tract Cancer (PRODIGE 12-ACCORD 18-UNICANCER GI): A Randomized Phase III Study. J Clin Oncol 2019;37:658-67. [Crossref] [PubMed]
- Chiang NJ, Chen LT. Adjuvant therapy in biliary tract cancers: oral fluoropyrimidine as a beginning. Lancet 2023;401:170-1. [Crossref] [PubMed]
- Ioka T, Shindo Y, Ueno M, et al. Current progress in perioperative chemotherapy for biliary tract cancer. Ann Gastroenterol Surg 2023;7:565-71. [Crossref] [PubMed]
- Ramaswamy A, Ostwal VS, Bhargava PG, et al. 99P Bevacizumab plus erlotinib maintenance versus observation in advanced biliary tract cancers: Results from the phase II component of a prospective integrated phase II-phase III parallel design superiority clinical trial (BEER-BTC study). Ann Oncol 2023;34:S217.
- Mohan A, Griffith KA, Wuchu F, et al. Devimistat in Combination with Gemcitabine and Cisplatin in Biliary Tract Cancer: Preclinical Evaluation and Phase Ib Multicenter Clinical Trial (BilT-04). Clin Cancer Res 2023;29:2394-400. [Crossref] [PubMed]
- Kefas J, Bridgewater J, Vogel A, et al. Adjuvant therapy of biliary tract cancers. Ther Adv Med Oncol 2023;15:17588359231163785. [Crossref] [PubMed]
- Yoo C, Lamarca A, Choi HJ, et al. Brightline-2: a phase IIa/IIb trial of brigimadlin (BI 907828) in advanced biliary tract cancer, pancreatic ductal adenocarcinoma or other solid tumors. Future Oncol 2024;20:1069-77. [Crossref] [PubMed]
- Sasaki T, Isayama H, Nakai Y, et al. A randomized phase II study of gemcitabine and S-1 combination therapy versus gemcitabine monotherapy for advanced biliary tract cancer. Cancer Chemother Pharmacol 2013;71:973-9. [Crossref] [PubMed]
- Valle J, Wasan H, Palmer DH, et al. Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. N Engl J Med 2010;362:1273-81. [Crossref] [PubMed]
- Gedela S, Munot P, Vaidyanathan A, et al. Gemcitabine, Cisplatin, and Nab-Paclitaxel as a First-Line Therapy for Advanced Biliary Tract Cancers. J Gastrointest Cancer 2024;55:263-9. [Crossref] [PubMed]
- Sharma A, Dwary AD, Mohanti BK, et al. Best supportive care compared with chemotherapy for unresectable gall bladder cancer: a randomized controlled study. J Clin Oncol 2010;28:4581-6. [Crossref] [PubMed]
- Oh DY, Ruth He A, Qin S, et al. Durvalumab plus Gemcitabine and Cisplatin in Advanced Biliary Tract Cancer. NEJM Evid 2022;1:EVIDoa2200015.
- Sanft T, Day A, Peterson L, et al. NCCN Guidelines® Insights: Survivorship, Version 1.2022. J Natl Compr Canc Netw 2022;20:1080-90. [Crossref] [PubMed]
- Duan J, Cui L, Zhao X, et al. Use of Immunotherapy With Programmed Cell Death 1 vs Programmed Cell Death Ligand 1 Inhibitors in Patients With Cancer: A Systematic Review and Meta-analysis. JAMA Oncol 2020;6:375-84. [Crossref] [PubMed]
- Morizane C, Okusaka T, Mizusawa J, et al. Combination gemcitabine plus S-1 versus gemcitabine plus cisplatin for advanced/recurrent biliary tract cancer: the FUGA-BT (JCOG1113) randomized phase III clinical trial. Ann Oncol 2019;30:1950-8. [Crossref] [PubMed]
- Ebata T, Hirano S, Konishi M, et al. Randomized clinical trial of adjuvant gemcitabine chemotherapy versus observation in resected bile duct cancer. Br J Surg 2018;105:192-202. [Crossref] [PubMed]
- André T, Tournigand C, Rosmorduc O, et al. Gemcitabine combined with oxaliplatin (GEMOX) in advanced biliary tract adenocarcinoma: a GERCOR study. Ann Oncol 2004;15:1339-43. [Crossref] [PubMed]
- André T, Reyes-Vidal JM, Fartoux L, et al. Gemcitabine and oxaliplatin in advanced biliary tract carcinoma: a phase II study. Br J Cancer 2008;99:862-7. [Crossref] [PubMed]
- Harder J, Riecken B, Kummer O, et al. Outpatient chemotherapy with gemcitabine and oxaliplatin in patients with biliary tract cancer. Br J Cancer 2006;95:848-52. [Crossref] [PubMed]
- Manzione L, Romano R, Germano D. Chemotherapy with gemcitabine and oxaliplatin in patients with advanced biliary tract cancer: a single-institution experience. Oncology 2007;73:311-5. [Crossref] [PubMed]
- Kim ST, Kang JH, Lee J, et al. Capecitabine plus oxaliplatin versus gemcitabine plus oxaliplatin as first-line therapy for advanced biliary tract cancers: a multicenter, open-label, randomized, phase III, noninferiority trial. Ann Oncol 2019;30:788-95. [Crossref] [PubMed]
- Sakai D, Kanai M, Kobayashi S, et al. Randomized phase III study of gemcitabine, cisplatin plus S-1 (GCS) versus gemcitabine, cisplatin (GC) for advanced biliary tract cancer (KHBO1401-MITSUBA). Ann Oncol 2018;29:viii205. [Crossref] [PubMed]
- Kelley RK, Ueno M, Yoo C, et al. Pembrolizumab in combination with gemcitabine and cisplatin compared with gemcitabine and cisplatin alone for patients with advanced biliary tract cancer (KEYNOTE-966): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2023;401:1853-65. [Crossref] [PubMed]
- Ellis H, Raghavan S, Wolpin BM, et al. How we treat advanced biliary tract cancers in the second-line setting. Clin Adv Hematol Oncol 2023;21:35-42.
- Lamarca A, Hubner RA, David Ryder W, et al. Second-line chemotherapy in advanced biliary cancer: a systematic review. Ann Oncol 2014;25:2328-38. [Crossref] [PubMed]
- Zhang D, Dorman K, Westphalen CB, et al. Unresectable biliary tract cancer: Current and future systemic therapy. Eur J Cancer 2024;203:114046. [Crossref] [PubMed]
- Shroff RT, Guthrie KA, Scott AJ, et al. SWOG 1815: A phase III randomized trial of gemcitabine, cisplatin, and nab-paclitaxel versus gemcitabine and cisplatin in newly diagnosed, advanced biliary tract cancers. J Clin Oncol 2023;41:LBA490. [Crossref] [PubMed]
- Hyung J, Kim I, Kim KP, et al. Treatment With Liposomal Irinotecan Plus Fluorouracil and Leucovorin for Patients With Previously Treated Metastatic Biliary Tract Cancer: The Phase 2b NIFTY Randomized Clinical Trial. JAMA Oncol 2023;9:692-699. [Crossref] [PubMed]
- Zhou N, Li X, Yang Y, et al. Sintilimab plus nab-paclitaxel as second-line treatment for advanced biliary tract cancer: study protocol for an investigator-initiated phase 2 trial (NapaSinti trial). BMC Cancer 2023;23:729. [Crossref] [PubMed]
- Jin S, Zhao R, Zhou C, et al. Feasibility and tolerability of sintilimab plus anlotinib as the second-line therapy for patients with advanced biliary tract cancers: An open-label, single-arm, phase II clinical trial. Int J Cancer 2023;152:1648-58. [Crossref] [PubMed]
- Sia D, Tovar V, Moeini A, et al. Intrahepatic cholangiocarcinoma: pathogenesis and rationale for molecular therapies. Oncogene 2013;32:4861-70. [Crossref] [PubMed]
- Lamarca A, Barriuso J, McNamara MG, et al. Molecular targeted therapies: Ready for "prime time" in biliary tract cancer. J Hepatol 2020;73:170-85. [Crossref] [PubMed]
- Walter D, Hartmann S, Waidmann O. Update on cholangiocarcinoma: potential impact of genomic studies on clinical management. Z Gastroenterol 2017;55:575-81. [Crossref] [PubMed]
- Okano N, Pirozzi A, Abidoye O, et al. Systemic therapy for pretreated advanced biliary tract cancer: past developments and recent advances. Jpn J Clin Oncol 2025;55:675-90. [Crossref] [PubMed]
- Lowery MA, Ptashkin R, Jordan E, et al. Comprehensive Molecular Profiling of Intrahepatic and Extrahepatic Cholangiocarcinomas: Potential Targets for Intervention. Clin Cancer Res 2018;24:4154-61. [Crossref] [PubMed]
- Goyal L, Kongpetch S, Crolley VE, et al. Targeting FGFR inhibition in cholangiocarcinoma. Cancer Treat Rev 2021;95:102170. [Crossref] [PubMed]
- LaPelusa M, Heumann T, Goff L, et al. Targeted therapies in advanced biliary tract cancers-a narrative review. Chin Clin Oncol 2023;12:14. [Crossref] [PubMed]
- Goyal L, Govindan A, Sheth RA, et al. Prognosis and Clinicopathologic Features of Patients With Advanced Stage Isocitrate Dehydrogenase (IDH) Mutant and IDH Wild-Type Intrahepatic Cholangiocarcinoma. Oncologist 2015;20:1019-27. [Crossref] [PubMed]
- Borger DR, Tanabe KK, Fan KC, et al. Frequent mutation of isocitrate dehydrogenase (IDH)1 and IDH2 in cholangiocarcinoma identified through broad-based tumor genotyping. Oncologist 2012;17:72-9. [Crossref] [PubMed]
- Lowery MA, Abou-Alfa GK, Burris HA, et al. Phase I study of AG-120, an IDH1 mutant enzyme inhibitor: Results from the cholangiocarcinoma dose escalation and expansion cohorts. J Clin Oncol 2017;35:4015.
- Saha SK, Gordan JD, Kleinstiver BP, et al. Isocitrate Dehydrogenase Mutations Confer Dasatinib Hypersensitivity and SRC Dependence in Intrahepatic Cholangiocarcinoma. Cancer Discov 2016;6:727-39. [Crossref] [PubMed]
- Zhu AX, Macarulla T, Javle MM, et al. Final Overall Survival Efficacy Results of Ivosidenib for Patients With Advanced Cholangiocarcinoma With IDH1 Mutation: The Phase 3 Randomized Clinical ClarIDHy Trial. JAMA Oncol 2021;7:1669-77. [Crossref] [PubMed]
- 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;21:796-807. [Crossref] [PubMed]
- Liu G, Chen T, Ding Z, et al. Inhibition of FGF-FGFR and VEGF-VEGFR signalling in cancer treatment. Cell Prolif 2021;54:e13009. [Crossref] [PubMed]
- Jones RL, Macarulla T, Charlson JA, et al. A phase Ib/II study of olutasidenib in patients with relapsed/refractory IDH1 mutant solid tumors: Safety and efficacy as single agent. J Clin Oncol 2020;38:e16643.
- Mellinghoff IK, Penas-Prado M, Peters KB, et al. Vorasidenib, a Dual Inhibitor of Mutant IDH1/2, in Recurrent or Progressive Glioma; Results of a First-in-Human Phase I Trial. Clin Cancer Res 2021;27:4491-9. [Crossref] [PubMed]
- Farha N, Dima D, Ullah F, et al. Precision Oncology Targets in Biliary Tract Cancer. Cancers (Basel) 2023;15:2105. [Crossref] [PubMed]
- Prete MG, Cammarota A, D'Alessio A, et al. Current options and future directions of systemic therapy for advanced biliary tract cancer. Explor Target Antitumor Ther 2021;2:416-33. [Crossref] [PubMed]
- Katoh M. Fibroblast growth factor receptors as treatment targets in clinical oncology. Nat Rev Clin Oncol 2019;16:105-22. [Crossref] [PubMed]
- Helsten T, Elkin S, Arthur E, et al. The FGFR Landscape in Cancer: Analysis of 4,853 Tumors by Next-Generation Sequencing. Clin Cancer Res 2016;22:259-67. [Crossref] [PubMed]
- Silverman IM, Hollebecque A, Friboulet L, et al. Clinicogenomic Analysis of FGFR2-Rearranged Cholangiocarcinoma Identifies Correlates of Response and Mechanisms of Resistance to Pemigatinib. Cancer Discov 2021;11:326-39. [Crossref] [PubMed]
- Cleary JM, Voss MH, Meric-Bernstam F, et al. Safety and efficacy of the selective FGFR inhibitor debio 1347 in phase I study patients with FGFR genomically activated advanced biliary tract cancer (BTC). J Clin Oncol 2018;36:447.
- Javle M, Kelley RK, Roychowdhury S, et al. Updated results from a phase II study of infigratinib (BGJ398), a selective pan-FGFR kinase inhibitor, in patients with previously treated advanced cholangiocarcinoma containing FGFR2 fusions. Ann Oncol 2018;29:viii720.
- 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;21:671-84. [Crossref] [PubMed]
- Goyal L, Meric-Bernstam F, Hollebecque A, et al. Updated results of the FOENIX-CCA2 trial: Efficacy and safety of futibatinib in intrahepatic cholangiocarcinoma (iCCA) harboring FGFR2 fusions/rearrangements. J Clin Oncol 2022;40:4009.
- Javle M, Roychowdhury S, Kelley RK, et al. Infigratinib (BGJ398) in previously treated patients with advanced or metastatic cholangiocarcinoma with FGFR2 fusions or rearrangements: mature results from a multicentre, open-label, single-arm, phase 2 study. Lancet Gastroenterol Hepatol 2021;6:803-15.
- Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol 2001;2:127-37. [Crossref] [PubMed]
- Javle M, Borad MJ, Azad NS, et al. Pertuzumab and trastuzumab for HER2-positive, metastatic biliary tract cancer (MyPathway): a multicentre, open-label, phase 2a, multiple basket study. Lancet Oncol 2021;22:1290-300. [Crossref] [PubMed]
- Lee CK, Chon HJ, Cheon J, et al. Trastuzumab plus FOLFOX for HER2-positive biliary tract cancer refractory to gemcitabine and cisplatin: a multi-institutional phase 2 trial of the Korean Cancer Study Group (KCSG-HB19-14). Lancet Gastroenterol Hepatol 2023;8:56-65. [Crossref] [PubMed]
- Ohba A, Morizane C, Ueno M, et al. Multicenter phase II trial of trastuzumab deruxtecan for HER2-positive unresectable or recurrent biliary tract cancer: HERB trial. Future Oncol 2022;18:2351-60. [Crossref] [PubMed]
- Terrell EM, Morrison DK. Ras-Mediated Activation of the Raf Family Kinases. Cold Spring Harb Perspect Med 2019;9:a033746. [Crossref] [PubMed]
- Nakamura Y, Mizuno N, Sunakawa Y, et al. Tucatinib and Trastuzumab for Previously Treated Human Epidermal Growth Factor Receptor 2-Positive Metastatic Biliary Tract Cancer (SGNTUC-019): A Phase II Basket Study. J Clin Oncol 2023;41:5569-78. [Crossref] [PubMed]
- Zeng W, Mao R, Zhang Z, et al. Combination Therapies for Advanced Biliary Tract Cancer. J Clin Transl Hepatol 2023;11:490-501. [Crossref] [PubMed]
- Simbolo M, Fassan M, Ruzzenente A, et al. Multigene mutational profiling of cholangiocarcinomas identifies actionable molecular subgroups. Oncotarget 2014;5:2839-52. [Crossref] [PubMed]
- Goeppert B, Frauenschuh L, Renner M, et al. BRAF V600E-specific immunohistochemistry reveals low mutation rates in biliary tract cancer and restriction to intrahepatic cholangiocarcinoma. Mod Pathol 2014;27:1028-34. [Crossref] [PubMed]
- Robertson S, Hyder O, Dodson R, et al. The frequency of KRAS and BRAF mutations in intrahepatic cholangiocarcinomas and their correlation with clinical outcome. Hum Pathol 2013;44:2768-73. [Crossref] [PubMed]
- Subbiah V, Puzanov I, Blay JY, et al. Pan-Cancer Efficacy of Vemurafenib in BRAF (V600)-Mutant Non-Melanoma Cancers. Cancer Discov 2020;10:657-63. [Crossref] [PubMed]
- Mie T, Sasaki T, Okamoto T, et al. Current Status of Targeted Therapy for Biliary Tract Cancer in the Era of Precision Medicine. Cancers (Basel) 2024;16:879. [Crossref] [PubMed]
- Gouda MA, Subbiah V. Expanding the Benefit: Dabrafenib/Trametinib as Tissue-Agnostic Therapy for BRAF V600E-Positive Adult and Pediatric Solid Tumors. Am Soc Clin Oncol Educ Book 2023;43:e404770. [Crossref] [PubMed]
- Amatu A, Sartore-Bianchi A, Bencardino K, et al. Tropomyosin receptor kinase (TRK) biology and the role of NTRK gene fusions in cancer. Ann Oncol 2019;30:viii5-viii15.
- Cocco E, Scaltriti M, Drilon A. NTRK fusion-positive cancers and TRK inhibitor therapy. Nat Rev Clin Oncol 2018;15:731-47. [Crossref] [PubMed]
- Vaishnavi A, Le AT, Doebele RC. TRKing down an old oncogene in a new era of targeted therapy. Cancer Discov 2015;5:25-34. [Crossref] [PubMed]
- Drilon A, Laetsch TW, Kummar S, et al. Efficacy of Larotrectinib in TRK Fusion-Positive Cancers in Adults and Children. N Engl J Med 2018;378:731-9. [Crossref] [PubMed]
- Tognon C, Knezevich SR, Huntsman D, et al. Expression of the ETV6-NTRK3 gene fusion as a primary event in human secretory breast carcinoma. Cancer Cell 2002;2:367-76. [Crossref] [PubMed]
- Solomon JP, Linkov I, Rosado A, et al. NTRK fusion detection across multiple assays and 33,997 cases: diagnostic implications and pitfalls. Mod Pathol 2020;33:38-46. [Crossref] [PubMed]
- Hong DS, DuBois SG, Kummar S, et al. Larotrectinib in patients with TRK fusion-positive solid tumours: a pooled analysis of three phase 1/2 clinical trials. Lancet Oncol 2020;21:531-40. [Crossref] [PubMed]
- Demols A, Rocq L, Perez-Casanova L, et al. A Two-Step Diagnostic Approach for NTRK Gene Fusion Detection in Biliary Tract and Pancreatic Adenocarcinomas. Oncologist 2023;28:e520-5. [Crossref] [PubMed]
- Perkhofer L, Beutel AK, Ettrich TJ. Immunotherapy: Pancreatic Cancer and Extrahepatic Biliary Tract Cancer. Visc Med 2019;35:28-37. [Crossref] [PubMed]
- Andrews LP, Yano H, Vignali DAA. Inhibitory receptors and ligands beyond PD-1, PD-L1 and CTLA-4: breakthroughs or backups. Nat Immunol 2019;20:1425-34. [Crossref] [PubMed]
- Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med 2015;372:320-30. [Crossref] [PubMed]
- Sharma P, Retz M, Siefker-Radtke A, et al. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial. Lancet Oncol 2017;18:312-22. [Crossref] [PubMed]
- El-Khoueiry AB, Sangro B, Yau T, et al. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 2017;389:2492-502. [Crossref] [PubMed]
- Casak SJ, Marcus L, Fashoyin-Aje L, et al. FDA Approval Summary: Pembrolizumab for the First-line Treatment of Patients with MSI-H/dMMR Advanced Unresectable or Metastatic Colorectal Carcinoma. Clin Cancer Res 2021;27:4680-4. [Crossref] [PubMed]
- Nakajima EC, Vellanki PJ, Larkins E, et al. FDA Approval Summary: Nivolumab in Combination with Ipilimumab for the Treatment of Unresectable Malignant Pleural Mesothelioma. Clin Cancer Res 2022;28:446-51. [Crossref] [PubMed]
- Zhao Y, Yang M, Feng J, et al. Advances in immunotherapy for biliary tract cancers. Chin Med J (Engl) 2024;137:524-32. [Crossref] [PubMed]
- 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;6:888-94. [Crossref] [PubMed]
- Zhang Z, Wang X, Li H, et al. Case Report: Camrelizumab combined with gemcitabine and oxaliplatin in the treatment of advanced intrahepatic cholangiocarcinoma: a case report and literature review. Front Immunol 2023;14:1230261. [Crossref] [PubMed]
- Sasaki T, Takeda T, Okamoto T, et al. Chemotherapy for Biliary Tract Cancer in 2021. J Clin Med 2021;10:3108. [Crossref] [PubMed]
- Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017;357:409-13. [Crossref] [PubMed]
- 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;147:2190-8. [Crossref] [PubMed]
- 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:e000367. [Crossref] [PubMed]
- Klein O, Kee D, Nagrial A, et al. Evaluation of Combination Nivolumab and Ipilimumab Immunotherapy in Patients With Advanced Biliary Tract Cancers: Subgroup Analysis of a Phase 2 Nonrandomized Clinical Trial. JAMA Oncol 2020;6:1405-9. [Crossref] [PubMed]
- Laface C, Fina E, Ricci AD, et al. Immunobiology of biliary tract cancer and recent clinical findings in approved and upcoming immune checkpoint inhibitors. Expert Opin Biol Ther 2024;24:1363-74. [Crossref] [PubMed]
- Morizane C, Ueno M, Ikeda M, et al. Update for: New developments in systemic therapy for advanced biliary tract cancer. Jpn J Clin Oncol 2025;55:210-8. [Crossref] [PubMed]
- Sung E, Ko M, Won JY, et al. LAG-3xPD-L1 bispecific antibody potentiates antitumor responses of T cells through dendritic cell activation. Mol Ther 2022;30:2800-16. [Crossref] [PubMed]
- Lan Y, Zhang D, Xu C, et al. Enhanced preclinical antitumor activity of M7824, a bifunctional fusion protein simultaneously targeting PD-L1 and TGF-β. Sci Transl Med 2018;10:eaan5488. [Crossref] [PubMed]
- Yoo C, Oh DY, Choi HJ, et al. Phase I study of bintrafusp alfa, a bifunctional fusion protein targeting TGF-β and PD-L1, in patients with pretreated biliary tract cancer. J Immunother Cancer 2020;8:e000564. [Crossref] [PubMed]
- Vitale E, Rizzo A, Maistrello L, et al. Sex differences in adverse events among cancer patients receiving immune checkpoint inhibitors: the MOUSEION-07 systematic review and meta-analysis. Sci Rep 2024;14:28309. [Crossref] [PubMed]
- Doki Y, Ueno M, Hsu CH, et al. Tolerability and efficacy of durvalumab, either as monotherapy or in combination with tremelimumab, in patients from Asia with advanced biliary tract, esophageal, or head-and-neck cancer. Cancer Med 2022;11:2550-60. [Crossref] [PubMed]
- Floudas CS, Xie C, Brar G, et al. Combined immune checkpoint inhibition (ICI) with tremelimumab and durvalumab in patients with advanced hepatocellular carcinoma (HCC) or biliary tract carcinomas (BTC). J Clin Oncol 2019;37:336.
- Oh DY, Lee KH, Lee DW, et al. Gemcitabine and cisplatin plus durvalumab with or without tremelimumab in chemotherapy-naive patients with advanced biliary tract cancer: an open-label, single-centre, phase 2 study. Lancet Gastroenterol Hepatol 2022;7:522-32. [Crossref] [PubMed]
- Oh DY, Lee KH, Lee DW, 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.
- Sahai V, Griffith KA, Beg MS, et al. A randomized phase 2 trial of nivolumab, gemcitabine, and cisplatin or nivolumab and ipilimumab in previously untreated advanced biliary cancer: BilT-01. Cancer 2022;128:3523-30. [Crossref] [PubMed]
- 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;4:611-21.
- Boilève A, Hilmi M, Gougis P, et al. Triplet combination of durvalumab, tremelimumab, and paclitaxel in biliary tract carcinomas: Safety run-in results of the randomized IMMUNOBIL PRODIGE 57 phase II trial. Eur J Cancer 2021;143:55-63. [Crossref] [PubMed]
- Arkenau HT, Martin-Liberal J, Calvo E, et al. Ramucirumab Plus Pembrolizumab in Patients with Previously Treated Advanced or Metastatic Biliary Tract Cancer: Nonrandomized, Open-Label, Phase I Trial (JVDF). Oncologist 2018;23:1407-e136. [Crossref] [PubMed]
- 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:e152670. [Crossref] [PubMed]
- 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 2020;9:414-24. [Crossref] [PubMed]
- Yang S, Zou R, Dai Y, et al. Tumor immune microenvironment and the current immunotherapy of cholangiocarcinoma Int J Oncol 2023;63:137. (Review). [Crossref] [PubMed]
- Rohaan MW, Wilgenhof S, Haanen JBAG. Adoptive cellular therapies: the current landscape. Virchows Arch 2019;474:449-61. [Crossref] [PubMed]
- Han S, Lee SY, Wang WW, et al. A Perspective on Cell Therapy and Cancer Vaccine in Biliary Tract Cancers (BTCs). Cancers (Basel) 2020;12:3404. [Crossref] [PubMed]
- Higuchi R, Yamamoto M, Hatori T, et al. Intrahepatic cholangiocarcinoma with lymph node metastasis successfully treated by immunotherapy with CD3-activated T cells and dendritic cells after surgery: report of a case. Surg Today 2006;36:559-62. [Crossref] [PubMed]
- Sharpe M, Mount N. Genetically modified T cells in cancer therapy: opportunities and challenges. Dis Model Mech 2015;8:337-50. [Crossref] [PubMed]
- Feng K, Liu Y, Guo Y, et al. Phase I study of chimeric antigen receptor modified T cells in treating HER2-positive advanced biliary tract cancers and pancreatic cancers. Protein Cell 2018;9:838-47. [Crossref] [PubMed]
- Guo Y, Feng K, Liu Y, et al. Phase I Study of Chimeric Antigen Receptor-Modified T Cells in Patients with EGFR-Positive Advanced Biliary Tract Cancers. Clin Cancer Res 2018;24:1277-86. [Crossref] [PubMed]
- Ivica NA, Young CM. Tracking the CAR-T Revolution: Analysis of Clinical Trials of CAR-T and TCR-T Therapies for the Treatment of Cancer (1997-2020). Healthcare (Basel) 2021;9:1062. [Crossref] [PubMed]
- Tran E, Turcotte S, Gros A, et al. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science 2014;344:641-5. [Crossref] [PubMed]
- Shimizu K, Kotera Y, Aruga A, et al. Clinical utilization of postoperative dendritic cell vaccine plus activated T-cell transfer in patients with intrahepatic cholangiocarcinoma. J Hepatobiliary Pancreat Sci 2012;19:171-8. [Crossref] [PubMed]
- Kou X, Yang XJ, Huang Y, et al. Autologous dendritic cells vaccine combined with cytokine-induced killer cells in treatment of biliary tract cancer: A clinical observation of efficacy. Academic Journal of Second Military Medical University 2015;36:897-900.
- Tao R, Krishnan S, Bhosale PR, et al. Ablative Radiotherapy Doses Lead to a Substantial Prolongation of Survival in Patients With Inoperable Intrahepatic Cholangiocarcinoma: A Retrospective Dose Response Analysis. J Clin Oncol 2016;34:219-26. [Crossref] [PubMed]
- Manterola C, Duque G, Grande L, et al. A systematic review of the effectiveness of adjuvant therapy for patients with gallbladder cancer. HPB (Oxford) 2019;21:1427-35. [Crossref] [PubMed]
- Ben-Josef E, Guthrie KA, El-Khoueiry AB, et al. SWOG S0809: A Phase II Intergroup Trial of Adjuvant Capecitabine and Gemcitabine Followed by Radiotherapy and Concurrent Capecitabine in Extrahepatic Cholangiocarcinoma and Gallbladder Carcinoma. J Clin Oncol 2015;33:2617-22. [Crossref] [PubMed]
- Shroff RT, Kennedy EB, Bachini M, et al. Adjuvant Therapy for Resected Biliary Tract Cancer: ASCO Clinical Practice Guideline. J Clin Oncol 2019;37:1015-27. [Crossref] [PubMed]
- Oneda E, Abu Hilal M, Zaniboni A. Biliary Tract Cancer: Current Medical Treatment Strategies. Cancers (Basel) 2020;12:1237. [Crossref] [PubMed]
- Scheuermann U, Kaths JM, Heise M, et al. Comparison of resection and transarterial chemoembolisation in the treatment of advanced intrahepatic cholangiocarcinoma--a single-center experience. Eur J Surg Oncol 2013;39:593-600. [Crossref] [PubMed]
- Herber S, Otto G, Schneider J, et al. Transarterial chemoembolization (TACE) for inoperable intrahepatic cholangiocarcinoma. Cardiovasc Intervent Radiol 2007;30:1156-65. [Crossref] [PubMed]
- Lin S, Hoffmann K, Schemmer P. Treatment of hepatocellular carcinoma: a systematic review. Liver Cancer 2012;1:144-58. [Crossref] [PubMed]
- Kishi Y, Shimada K, Nara S, et al. Role of hepatectomy for recurrent or initially unresectable hepatocellular carcinoma. World J Hepatol 2014;6:836-43. [Crossref] [PubMed]
- Lencioni R. Loco-regional treatment of hepatocellular carcinoma. Hepatology 2010;52:762-73. [Crossref] [PubMed]
- Wang Z, Ren Z, Chen Y, et al. Adjuvant Transarterial Chemoembolization for HBV-Related Hepatocellular Carcinoma After Resection: A Randomized Controlled Study. Clin Cancer Res 2018;24:2074-81. [Crossref] [PubMed]
- Kudo M, Ueshima K, Ikeda M, et al. Final Results of TACTICS: A Randomized, Prospective Trial Comparing Transarterial Chemoembolization Plus Sorafenib to Transarterial Chemoembolization Alone in Patients with Unresectable Hepatocellular Carcinoma. Liver Cancer 2022;11:354-67. [Crossref] [PubMed]
- Peng Z, Fan W, Zhu B, et al. Lenvatinib Combined With Transarterial Chemoembolization as First-Line Treatment for Advanced Hepatocellular Carcinoma: A Phase III, Randomized Clinical Trial (LAUNCH). J Clin Oncol 2023;41:117-27. [Crossref] [PubMed]
- Bester L, Meteling B, Boshell D, et al. Transarterial chemoembolisation and radioembolisation for the treatment of primary liver cancer and secondary liver cancer: a review of the literature. J Med Imaging Radiat Oncol 2014;58:341-52. [Crossref] [PubMed]
- Schönherr J, Seifert P, Gühne F, et al. Transarterial Radioembolization (TARE) in Patients with Hepatocellular Carcinoma: A Comparison of Palliative with Bridging-to-Transplant Concepts. Cancers (Basel) 2024;16:235. [Crossref] [PubMed]
- Aliseda D, Martí-Cruchaga P, Zozaya G, et al. Liver Resection and Transplantation Following Yttrium-90 Radioembolization for Primary Malignant Liver Tumors: A 15-Year Single-Center Experience. Cancers (Basel) 2023;15:733. [Crossref] [PubMed]
- Lopez-Lopez V, Miura K, Kuemmerli C, et al. Selecting the Appropriate Downstaging and Bridging Therapies for Hepatocellular Carcinoma: What Is the Role of Transarterial Radioembolization? A Pooled Analysis. Cancers (Basel) 2023;15:2122. [Crossref] [PubMed]
- Hickey R, Mulcahy MF, Lewandowski RJ, et al. Chemoradiation of hepatic malignancies: prospective, phase 1 study of full-dose capecitabine with escalating doses of yttrium-90 radioembolization. Int J Radiat Oncol Biol Phys 2014;88:1025-31. [Crossref] [PubMed]
- Stecca T, Greco A, Brollo PP, et al. Hepatic arterial infusion chemotherapy: a review with technical notes. Hepatoma Res 2024;10:28.
- Thiels CA, D'Angelica MI. Hepatic artery infusion pumps. J Surg Oncol 2020;122:70-7. [Crossref] [PubMed]
- Kemeny NE, Niedzwiecki D, Hollis DR, et al. Hepatic arterial infusion versus systemic therapy for hepatic metastases from colorectal cancer: a randomized trial of efficacy, quality of life, and molecular markers (CALGB 9481). J Clin Oncol 2006;24:1395-403. [Crossref] [PubMed]
- Massani M, Bonariol L, Stecca T. Hepatic Arterial Infusion Chemotherapy for Unresectable Intrahepatic Cholangiocarcinoma, a Comprehensive Review. J Clin Med 2021;10:2552. [Crossref] [PubMed]
- Jarnagin WR, Schwartz LH, Gultekin DH, et al. Regional chemotherapy for unresectable primary liver cancer: results of a phase II clinical trial and assessment of DCE-MRI as a biomarker of survival. Ann Oncol 2009;20:1589-95. [Crossref] [PubMed]
- Kemeny NE, Schwartz L, Gönen M, et al. Treating primary liver cancer with hepatic arterial infusion of floxuridine and dexamethasone: does the addition of systemic bevacizumab improve results? Oncology 2011;80:153-9. [Crossref] [PubMed]
- Ghiringhelli F, Lorgis V, Vincent J, et al. Hepatic arterial infusion of gemcitabine plus oxaliplatin as second-line treatment for locally advanced intrahepatic cholangiocarcinoma: preliminary experience. Chemotherapy 2013;59:354-60. [Crossref] [PubMed]
- Konstantinidis IT, Groot Koerkamp B, Do RK, et al. Unresectable intrahepatic cholangiocarcinoma: Systemic plus hepatic arterial infusion chemotherapy is associated with longer survival in comparison with systemic chemotherapy alone. Cancer 2016;122:758-65. [Crossref] [PubMed]
- Cercek A, Boerner T, Tan BR, et al. Assessment of Hepatic Arterial Infusion of Floxuridine in Combination With Systemic Gemcitabine and Oxaliplatin in Patients With Unresectable Intrahepatic Cholangiocarcinoma: A Phase 2 Clinical Trial. JAMA Oncol 2020;6:60-7. [Crossref] [PubMed]
- Chu KF, Dupuy DE. Thermal ablation of tumours: biological mechanisms and advances in therapy. Nat Rev Cancer 2014;14:199-208. [Crossref] [PubMed]
- Brace C. Thermal tumor ablation in clinical use. IEEE Pulse 2011;2:28-38. [Crossref] [PubMed]
(English Language Editor: J. Gray)

