Next Article in Journal
Clinical Effects of RUNX1 Mutations on the Outcomes of Patients with Acute Myeloid Leukemia Treated with Allogeneic Hematopoietic Stem-Cell Transplantation
Previous Article in Journal
Exploring Cancer Patients’ and Caregivers’ Perspectives and Knowledge Regarding Biomarker Testing in Canada
Previous Article in Special Issue
A New Prognostic Indicator for Biliary Tract Cancers: The ABIC Score
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Advances in Initially Unresectable Locally Advanced Intrahepatic Cholangiocarcinoma: Emerging Treatments and the Role of Liver Transplantation

Division of Transplant and HPB Surgery, Department of Surgery, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
*
Author to whom correspondence should be addressed.
Curr. Oncol. 2025, 32(6), 293; https://doi.org/10.3390/curroncol32060293
Submission received: 5 April 2025 / Revised: 9 May 2025 / Accepted: 19 May 2025 / Published: 22 May 2025
(This article belongs to the Special Issue Biliary Tract Cancer Updates: Advancements and Insights)

Abstract

:
Optimal curative therapy for intrahepatic cholangiocarcinoma (iCCA) involves hepatic resection; however, due to its insidious nature, iCCA frequently presents at advanced stages. Consequently, 70–80% of patients feature unresectable iCCA at presentation. Recent expansions in therapeutic options for locally advanced unresectable iCCA include immunotherapy, targeted chemotherapeutics, and liver-directed therapies. These have increased progression-free survival, enhanced response rates, and improved downstaging for resection. Liver transplant has also emerged as an alternative for patients whose tumors remain unresectable despite therapeutic response. Here, we explore emerging treatment options included in a multidisciplinary treatment paradigm to prolong survival in patients with initially unresectable locally advanced iCCA.

1. Introduction

Cholangiocarcinoma is a highly aggressive malignancy arising from the biliary epithelium, and it is classified according to its anatomic site of origin [1]. Intrahepatic cholangiocarcinoma (iCCA) arises from segmental and distal ducts within the liver parenchyma. Extrahepatic cholangiocarcinoma subtypes include perihilar cholangiocarcinoma (pCCA), originating from the right, left, and/or common hepatic duct, and distal cholangiocarcinoma (dCCA), originating from the common bile duct distal to the insertion of the cystic duct [2,3]. Intrahepatic CCA is further classified by a growth pattern into mass-forming, periductal infiltrating, and intraductal subtypes [3]. Mass-forming iCCA is the most common, with a prevalence of 60%, while 20% of cases are classified as either periductal or mixed type [4].
CCA accounts for 10–15% of liver cancers, and iCCA is the least common subtype (10–20% are CCA) [5]. The incidence of iCCA in the United States, however, has steadily increased from 0.49 to 1.48 per 100,000 persons from 2000 to 2019 [6]. Median overall survival (OS) has recently improved but remains dismal (from 5 months in 2000 to 9 months in 2017) [6]. The best outcomes for iCCA are achieved with resection in the absence of distant lymph nodes or metastatic disease, and long-term survival for patients with initially unresectable locally advanced disease has been historically limited. In the present review, we explore emerging treatment options included in a multidisciplinary treatment paradigm to prolong survival in patients with initially unresectable locally advanced iCCA.

2. Pathogenesis, Presentation, and Diagnosis

Risk factors for iCCA often relate to biliary inflammation, which includes parasitic infection, choledochal cyst, chronic biliary disease, recurrent pyogenic cholangitis, hepatitis B or C, liver flukes, and toxin exposure (asbestos, dioxins, or nitrosamines) [2,6]. Metabolic diseases such as diabetes, obesity, metabolic dysfunction associated with steatotic liver disease (MASLD), and cirrhosis also increase the risk, although iCCA can arise in either cirrhotic or non-cirrhotic livers [2].
Diagnosis of iCCA often occurs at late stages due to its lack of early symptoms and limited detection through non-invasive methods [5,7]. While pCCA and dCCA often present with jaundice due to biliary obstruction, iCCA rarely presents with jaundice [7]. At advanced stages, iCCA presents with non-specific symptoms, including abdominal pain, malaise, nausea, anorexia, and weight loss [7]. Early-stage diagnosis is usually incidental (20–25%) during imaging performed for screening or other indications [7]. Blood biomarkers, including CA19-9 and CEA, can be evaluated, but their specificity is limited as benign liver pathologies may also increase biomarker levels [4].
High-quality cross-sectional imaging, such as contrast-enhanced multiphasic CT, MRI, and MRCP, is necessary for the staging and assessment of primary tumor location, vascular invasion, lymph node involvement, and distant metastases [7]. On CT, iCCA appears as a well-defined or infiltrative hypodense hepatic lesion, which may demonstrate distal biliary dilatation or capsular retraction [4]. Due to its portal blood supply, portal-phase or delayed-phase enhancement may distinguish it from hepatocellular carcinoma (HCC). The mass may also demonstrate peripheral arterial hyperenhancement, with centripetal enhancement in later phases [8]. On MRI or MRCP, iCCAs appear as hypointense lesions on T1- and heterogeneously hyperintense lesions on T2-weighted images [4]. The lesion may demonstrate peripherally restricted diffusion, with less restriction in central areas. Gadoxetic acid-enhanced MRI may distinguish between mixed HCC-CCA and mass-forming iCCA, as iCCA may demonstrate a target-shaped enhancement [4,8].
Unfortunately, the sensitivity of MRI or CT alone in the detection of distant lymph node metastases is quite low. While FDG-PET is not routinely used to stage iCCA, it may be useful to identify occult metastases. This may inform candidacy for resection or transplant [4,8]. EUS/ERCP may supplement evaluation for intervention on biliary obstruction and EUS-guided aspiration of enlarged lymph nodes. Ultimately, diagnostic laparoscopy is recommended for evaluation of peritoneal and lymph node involvement prior to surgical intervention.
Histopathologic or cytologic analysis is generally required to confirm the diagnosis. Brush cytology or biopsy can be performed using ERCP or PTC (diagnostic sensitivities of 56% for brushings, 67% for biopsy, and 70.7% for biopsy+brushings). Alternatively, EUS-guided fine needle aspiration (FNA) can be employed; however, it is more effective for extrahepatic CCA, and concern for peritoneal seeding limits its utility [4]. For iCCA, image-guided core needle biopsy is preferred, as it provides a larger diagnostic sample sufficient for additional genetic analysis [9]. For all biopsy samples, next-generation sequencing should be performed to assess eligibility for immunotherapy and targeted systemic therapy options.

3. Surgery as the Gold Standard for iCCA Treatment

Resection with negative microscopic margins is the only proven curative option for iCCA. Unfortunately, only a third of patients presenting with iCCA are eligible for resection [10]. Five-year overall survival (OS) following resection is poor, at 20–35%, with recurrence-free survival (RFS) at 2–39%, with a median OS of 28 months [1,11]. While tumor number, vascular invasion, and lymph node involvement are prognostic indicators for patients following resection, tumor size is not a significant prognostic factor [12]. Prognosis following R0 resection differs based on TNM stages, with a 5-year OS of 45.3%, 28.9%, 13.7%, and 0% for TNM stages I, II, III, and IV, respectively [13]. Thus, for patients with locally advanced or initially unresectable iCCA, alternative or combined treatment modalities should be considered.
Patients whose tumors can be resected with negative histologic margins while maintaining an adequate liver remnant should be offered upfront resection. An adequate future liver remnant (FLR) requires a minimum of two contiguous segments with sufficient perfusion and both venous and biliary drainage. Underlying parenchymal dysfunction further defines the required volume and is estimated at 20% of the total liver volume for a normal liver, 30% for an injured one, and ≥40% for fibrotic or cirrhotic liver. Approaches to increase the FLR include portal vein embolization, combined portal and hepatic vein embolization, and Y90. Extrahepatic disease (including lymph nodes beyond the regional basin) is a contraindication to resection. Bilateral multifocal or multicentric disease also has poor outcomes following resection [9]. Evidence supporting the survival benefit of R1 resections is lacking [14,15]. Data is emerging to suggest that initially unresectable iCCA, especially in the setting of liver-limited disease, may be downstaged to allow for resection.
Adjuvant therapy following curative intent resection in iCCA has proven efficacy. The BILCAP trial evaluated oral capecitabine following biliary tract cancer (BTC) resection, demonstrating a median OS of 51.1 months with adjuvant capecitabine compared to 36.4 months in observation. RFS was also improved to 24.4 months with capecitabine versus 17.5 months in observation [16]. Trials evaluating alternative adjuvant regimens include the PRODIGE 12 trial. Patients were randomized to either GEMOX (gemcitabine + cisplatin) or surveillance following R0 or R1 resection for BTC. RFS with GEMOX was improved to 30.4 months compared to 18.5 months with observation, but the results were not statistically significant [17]. The ESPAC-3 reported a statistically significant OS benefit with adjuvant fluorouracil or gemcitabine following resection for pancreatic and BTC [18]. Based on the findings from the BILCAP trial, the ASCO clinical practice guideline currently recommends adjuvant capecitabine for 6 months following BTC resection [19].

4. Systemic Chemotherapy for Initially Unresectable iCCA

First-line treatment for locally advanced and unresectable iCCA is systemic chemotherapy, which can downstage for resection, treat micrometastatic disease, and halt disease progression. The ABC-02 trial established a significant survival benefit to using gemcitabine and cisplatin (Gem/Cis) combination therapy for advanced BTC, leading to its adoption as the standard first-line regimen [20]. Gem/Cis remained the standard treatment until the publication of the TOPAZ-1 trial in 2022. TOPAZ-1 evaluated durvalumab (a monoclonal antibody against PD-L1) plus Gem/Cis (Gem/Cis/Durva) versus Gem/Cis+placebo, in which 685 patients with unresectable, metastatic, or recurrent BTC were randomized. Twenty-four-month OS with Gem/Cis/Durva was 24.9% compared with 10.4% for Gem/Cis alone [21]. Pembrolizumab (a monoclonal antibody against PD-1) in combination with Gem/Cis (Gem/Cis/Pembro) was assessed in the KEYNOTE-966 trial. Median OS with Gem/Cis/Pembro was 12.7 months compared with 10.9 months for Gem/Cis alone (p = 0.0034) with similar toxicity [22]. This evidence supports adding immunotherapy to Gem/Cis in the first-line setting. An alternative combination of Gem/Cis and nab-paclitaxel (GAP) has been concurrently evaluated for systemic treatment for advanced BTC. Significant enthusiasm for the combination arose from the single-arm, phase 2 results, which demonstrated improved progression-free survival (PFS) (11.8 months vs. 8 months) and prolonged OS (19.2 months vs. 11.2 months) compared to historical Gem/Cis controls [23]. This prompted the phase III SWOG1815 trial, which randomized 441 advanced BTC patients to either GAP or standard Gem/Cis dosing. While there was no overall statistically significant improvement in OS (14 months vs. 12.7 months, respectively), select patient groups, including those with locally advanced disease, did exhibit some benefit [24].
Neoadjuvant for downstaging: With improved disease response observed in TOPAZ and SWOG for patients with locally advanced iCCA, there has been a resurgent interest in neoadjuvant chemotherapy for downstaging prior to liver resection. Neoadjuvant chemotherapy has the potential to downstage tumors, control micrometastatic disease, eradicate lymph node metastases, and improve the probability of achieving R0 resection [25]. Current evidence regarding the use of neoadjuvant treatment largely comes from retrospective and single-case analyses [26]. A propensity-matched retrospective analysis of 1450 patients demonstrated superior OS with neoadjuvant versus adjuvant chemotherapy [25]. Another single-institution retrospective analysis demonstrated prolonged OS with resection with or without neoadjuvant therapy for iCCA compared to chemotherapy alone (24.1, 25.7, and 7.8 months, respectively). While neoadjuvant therapy did not significantly prolong OS compared to surgery alone, 39 of 74 patients receiving neoadjuvant therapy had initially unresectable locally advanced iCCA. Thus, initially unresectable patients who are successfully downstaged demonstrate similar OS and RFS to patients whose tumor is initially resectable [27].
Prospective studies evaluating neoadjuvant therapies for cholangiocarcinoma are underway. A recent multi-institutional, phase II trial evaluating neoadjuvant gemcitabine, cisplatin, and nab-paclitaxel (NEO-GAP) for resectable high-risk intrahepatic cholangiocarcinoma demonstrated that this strategy is feasible and not associated with significant postoperative morbidity. All 30 enrolled patients completed preoperative chemotherapy, and 22 patients completed resection per protocol. Of 30 enrolled patients, 90% exhibited disease control, with 23% demonstrating a partial response. A total of 73% of patients completed therapy and progressed to resection. Median OS for the entire cohort was 24 months, with recurrence in 43% of patients following surgery [28]. Thus, neoadjuvant regimens may improve outcomes for patients with initially unresectable disease.
Given the demonstrated feasibility, safety, and efficacy of neoadjuvant treatment in the NEO-GAP trial, additional neoadjuvant trials evaluating combined chemotherapy and immunotherapy or targeted therapy have been initiated. Building off the NEO-GAP study, the upcoming OPT-IC trial (NCT03579771) will perform a phase II feasibility and safety assessment of neoadjuvant gemcitabine, cisplatin, and nab-paclitaxel, with an FGFR2 inhibitor added for patients with FGFR2 fusion or rearrangements [29]. The NCI’s Experimental Therapeutics Clinical Trials Network (ETCTN) is conducting a phase II trial evaluating neoadjuvant durvalumab with gemcitabine/cisplatin chemotherapy in patients with high-risk, resectable iCCA (ETCTN-10608, NCT06050252). Primary objectives are evaluation of treatment drop-out and completion of therapy/resection, with a secondary objective of determining pathologic response [30]. MD Anderson Cancer Center is similarly conducting a single-arm, phase II trial assessing pembrolizumab in combination with gemcitabine/cisplatin for borderline resectable iCCA, with RFS and major pathologic response as primary endpoints (NCT05967182) [31]. Finally, the NEOLANGIO trial (NCT06569225) at the University of Toronto will evaluate neoadjuvant gemcitabine, cisplatin, and nab-paclitaxel, along with rilvegostomig (an anti-TGIT/anti-PD-1 bispecific antibody) in resectable iCCA [32]. Collectively, these efforts signal a growing interest in optimizing neoadjuvant strategies through the incorporation of immunotherapy, with the goal of enhancing surgical outcomes and overall survival. Prospective trials evaluating neoadjuvant regimens for iCCA are summarized in Table 1.
Targeted therapies: Cholangiocarcinoma exhibits substantial genetic heterogeneity, making next-generation sequencing valuable for identifying potential drug targets and predicting prognosis [33]. The most common mutations include IDH1, ARID1A, BAP1, TP53, and FGFR2 gene fusions [33]. FGFR2 gene arrangements and gain-of-function mutations in IDH1 are actionable through targeted treatment. Such targets are the subject of extensive clinical investigation as first- and second-line therapy [33]. Among cholangiocarcinoma patients with FGFR fusions or rearrangements, 35.5% achieved an objective response when treated with second-line pemigatinib, an oral inhibitor of FGFR1, FGFR2, and FGFR3 [34]. FGFR2 fusion- or rearrangement-positive cholangiocarcinoma patients treated with futibatinib, a covalent FGFR inhibitor, also experience clinical benefit [35]. Ivosidenib, an IDH-1 inhibitor, demonstrates prolonged OS and PFS in patients with IDH-1 mutant unresectable disease [20,36]. These findings highlight the clinical importance of genetic profiling to identify actionable mutations, and indicate that targeted therapies may offer future benefit for downstaging patients with locally advanced unresectable disease.

5. Use of Locoregional and Liver-Directed Therapies in Initially Unresectable iCCA

Locoregional therapy is a cornerstone of treatment in the setting of unresectable HCC, and such liver-directed options may add benefit to the treatment of locally advanced iCCA.
Radiation therapy: Advancements in radiation delivery and safety have driven interest in its use for advanced iCCA [37]. In a single-institution retrospective study, patients with inoperable iCCA treated with high-dose radiation demonstrated survival rates comparable to those previously reported with resection [38]. Proton beam therapy, a form of external beam radiation therapy (EBRT), has also shown promise. In a phase II clinical trial, treatment with high-dose hypofractionated proton therapy improved tumor control and survival in patients with unresectable iCCA [37]. Proton beam therapy is hypothesized to lessen unwanted radiation exposure and reduce the risk of radiation-induced liver disease [37]. Pencil beam scanning (PBS) proton beam therapy further decreases doses to nearby organs and reduces radiation-induced injury [39]. Proton beam therapy administered with PBS has been shown to be safe and effective on retrospective analysis for large and multifocal liver tumors, but further investigation is needed to confirm survival benefits [39]. High-dose iCCA radiation alone may be sufficient to improve OS and PFS; however, utility for downstaging, especially at high doses, may complicate and increase the morbidity of operative resection.
TACE: Transarterial chemoembolization (TACE) directly infuses a concentrated chemotherapeutic emulsion and iodized oil via the hepatic arterial system directly into tumor tissue. DEB-TACE delivers chemotherapy using drug-eluting beads rather than iodized oil. Localized delivery of chemotherapeutic agents minimizes hepatic and systemic toxicity [40]. While TACE has been extensively studied in HCC, evidence supporting its use for iCCA is primarily based on retrospective studies, particularly in unresectable disease [41]. For unresectable iCCA, retrospective analysis reveals prolonged overall survival with TACE [42,43,44,45]. Multicenter prospective phase II evaluation demonstrates improved OS in unresectable iCCA following combined drug-eluting irinotecan (DEBIRI) TACE combined with systemic Gem/Cis compared to Gem/Cis alone (33.7 vs. 12.6 months). Combined DEBIRI treatment additionally resulted in greater downstaging (25%) compared to Gem/Cis (8%) [45]. Evaluating prognostic factors, including tumor vascularity, initial response to TACE, and Child–Pugh classification, is important before initiating TACE to optimize patient outcomes [44].
TARE: Transarterial radioembolization (TARE) is gaining recognition as a valuable palliative and downstaging option for unresectable iCCA. Transarterial Y90 radioembolization uses beta-emitting microspheres to deliver selective intratumor radiation via hepatic arterial access [46]. A systematic review encompassing 21 predominantly retrospective studies evaluated demonstrated an PFS of 7.8 months and a pooled OS of 12.7 months with TARE [47]. The MISPHEC trial was the first published prospective trial investigating the efficacy of TARE combination with chemotherapy in unresectable iCCA. In 41 patients receiving first-line TARE+Gem/Cis, median OS was 22 months, and PFS was 55% at 12 months and 30% at 24 months [46]. Nine patients (22%) were successfully downstaged to surgical intervention, and eight patients (20%) achieved R0 resection [46]. The sequence of administration of chemotherapy and TARE remains a subject for investigation. In a phase 2 single-arm multicenter study evaluating TARE followed by Gem/Cis, investigators hypothesized that TARE followed by chemotherapy would enhance local control of iCCA prior to systemic control. Among the 16 patients treated, median OS was 21.6 months and median PFS was 9 months. While both clinical trials report a survival benefit to treating with a combination of SIRT and chemotherapy, further investigation is warranted to determine the optimal timing for treatment administration [48].
Hepatic artery infusion pump: Hepatic artery infusion (HAI) therapy delivers high doses of chemotherapy into hepatic arterial circulation using an implanted pump (generally via the gastroduodenal artery). This approach leverages hepatic first-pass metabolism to minimize systemic exposure while maximizing drug delivery to the liver [49,50]. In a comparative meta-analysis, HAI provided superior tumor response and OS compared to TACE, DEB-TACE, and TARE [51]. Retrospective data suggest that a combination of systemic therapy and HAI improves survival compared to systemic chemotherapy alone [52], and current research focuses on the efficacy of combining floxuridine HAI with systemic chemotherapy to maximize tumor response. An MSKCC single-institution phase 2 clinical trial of 38 patients with unresectable iCCA treated with HAI floxuridine plus systemic Gem/Ox demonstrated a PFS of 80% at 6 months with 84% disease control and 58% partial radiographic response at 6 months. Four patients were downstaged to resection. Interestingly, survival benefit was greatest in patients with IDH1/2 mutant tumors (2-year OS 90% vs. 33% for wild-type) [53]. Despite these promising results, large-scale randomized prospective studies to ascertain the true efficacy of HAI therapy are necessary.
Novel chemotherapy combinations with HAI are additionally being investigated. Sun Yat-Sen University Cancer Center (SYSUCC) recently reported that lenvatinib plus durvalumab combined with FOLFOX-HAI achieved a median OS of 17.9 months and a median PFS of 11.9 months, but 46% of patients experienced grade 3 or 4 complications [54]. HELIX-1 (NCT04251714) is an active, single-center, phase II clinical trial investigating the efficacy of the induction of systemic mFOLFIRINOX (folinic acid, fluorouracil, irinotecan, and oxaliplatin) followed by a concurrent HAI of floxuridine and dexamethasone plus systemic mFOLFIRI (folinic acid, fluorouracil, and irinotecan) for unresectable iCCA. Preliminary data presented at ASCO 2024 reported that the regimen is well-tolerated and demonstrates longer disease control compared to historical controls [55]. Future directions include exploring innovative chemotherapy combinations for HAI, conducting phase III trials to evaluate efficacy, and examining the impact of specific disease mutations on treatment outcomes.
Future therapies: While liver-directed therapies have demonstrated disease control and response, combination with emerging systemic treatments may represent the future for iCCA treatment. Recently, a phase II trial reported that external radiation combined with anti-PD1 antibodies for unresectable iCCA is safe and efficacious [56]. Other treatments under current investigation include IL-12 and FUDR HAI in combination with Gem/Ox for unresectable iCCA (NCT05286814) [57]. Finally, histotripsy, a non-invasive non-ionizing and non-thermal ultrasound-based ablation device, has recently been granted FDA clearance. Early studies demonstrate successful ablation of intrahepatic cholangiocarcinoma [58] and, theoretically, the release of tumor antigens through this ablation modality in conjunction with systemic immunotherapy may facilitate immune clearance of tumor cells. These promising results highlight the emerging role of combined liver-directed and systemic therapies in the treatment or downstaging of locally advanced iCCA. OS and PFS for prospective trials evaluating liver-directed therapies are summarized in Table 2.

6. Liver Transplantation for Intrahepatic Cholangiocarcinoma

Liver transplant (LT) for unresectable, locally advanced iCCA affords wider surgical margins than resection and treats underlying liver disease [59]. Despite this, LT for iCCA has been historically contraindicated due to poor survival outcomes and high recurrence [60]. A retrospective analysis conducted in 1991 reported 44% recurrence, with a 2- and 5-year OS of 30% and 17% in transplanted iCCA patients [61]. Failure to differentiate between cholangiocarcinoma subtypes, poor patient selection, and lack of neoadjuvant treatment could reduce the applicability of historic outcomes to current practice [62]. Interest in LT for iCCA has expanded with the recent introduction of revised protocols and improved patient selection.
Due to its long-standing contraindication, LT for iCCA studies have largely been retrospective, on patients misdiagnosed with HCC or on tumors discovered incidentally on explant [62]. Several more contemporary retrospective multicenter studies have compared survival and recurrence rates in cirrhotic LT recipients diagnosed with iCCA on explant. In these studies, patients with prior systemic chemotherapy were excluded, and all patients were either untreated or received only locoregional therapy. The first Spanish multicenter study included 29 cirrhotic patients with iCCA, eight of whom had “very early” iCCA (single tumor ≤ 2 cm). While patients with tumors ≥ 2 cm or multinodular tumors had recurrence rates of 36.4%, no patients with “very early” iCCA had a recurrence, and 1-, 3-, and 5-year OS was 100%, 73%, and 73%, respectively [63]. These findings were validated in 2016 through a multinational study by the same group, which reported decreased post-LT recurrence at 1, 3, and 5 years in “very early” iCCA patients (7%, 18%, and 18%) compared with 30%, 47%, and 61% for patients with “advanced disease” [64]. The “very early” iCCA group additionally showed an improved 1-, 3-, and 5-year OS of 93%, 84%, and 65%, compared to 79%, 50%, and 45% for those with “advanced disease” [64]. While these findings were promising, identification of iCCA patients with tumors ≤ 2 cm is rare, given the insidious nature of the disease. A French multicenter retrospective study subsequently compared the outcomes of LT and resection for cirrhotic patients with iCCA or combined HCC/CCA [65]. Overall, LT recipients had an improved 5-year RFS, at 75% compared to 36% for resection. On subgroup analysis, transplanted patients with tumors 2–5 cm had a 21% recurrence rate compared with 48% for those following resection, and a 5-year RFS of 74% compared with 40% for those following resection [65]. These findings suggest LT may be a viable option for cirrhotic patients with tumors ≤ 5 cm.
Due to the rarity of transplant for iCCA, results of retrospective studies have often combined data with the outcomes for hepatocholangiocarcinoma (HCC-CCA). Unlike iCCA, HCC-CCA is thought to arise from hepatic progenitor cells. While both tumor pathologies have been classified as aggressive, HCC-CCA has a distinctly different pathology. These tumors often present in the setting of pre-existing cirrhosis, making tumor resection difficult [66,67,68]. Up to 3% of tumors initially diagnosed as HCC are identified as HCC-CCA on explant. A few studies have separately considered LT outcomes for iCCA. One single-center analysis from UCLA performed propensity matching of patients diagnosed with HCC-CCA (n = 12) to patients with HCC (n = 36). HCC-CCA tumors were more likely to be poorly differentiated with a higher grade. When matched by explant pathologic criteria (diameter, differentiation, grade, vascular invasion, etc.), HCC-CCA and HCC LT recipients exhibited a comparable 5-year OS and RFS (42% vs. 48% and 42% vs. 44%, respectively), with recurrence limited to patients with poorly differentiated tumors [68]. Data from 12 U.S. transplant centers were subsequently analyzed for patients diagnosed with HCC-CCA compared to those with HCC. Patients with HCC-CCA meeting Milan criteria exhibited higher recurrence (23.1%) compared to those with HCC (11.5%), but 5-year OS was comparable (70.1% HCC-CCA, 73.4% HCC, p = 0.806). Given the different tumor biology, results for HCC-CCA should not be combined with those for iCCA as they risk an inadvertent bias of outcomes [69].
Improved survival outcomes for LT in iCCA patients with low tumor burden highlighted the need to clarify the role of neoadjuvant therapy in facilitating LT in locally advanced iCCA. Case series combining neoadjuvant treatment with transplantation (with or without cirrhosis) have also demonstrated a survival advantage in advanced disease. UCLA performed a retrospective analysis comparing LT and resection outcomes for iCCA and pCCA. The 5-year RFS was significantly higher for patients receiving transplant. In the LT group, neoadjuvant plus adjuvant therapy prolonged 5-year RFS to 47%, compared with 20% for no therapy, and 33% for adjuvant therapy alone [70].
Houston Methodist and MD Anderson Cancer Center subsequently led the first prospective case series evaluating neoadjuvant therapy and LT for locally advanced unresectable iCCA. Patients were required to demonstrate >6 months of disease stability on therapy (primarily Gem/Cis) with no restrictions on tumor number or size. The initial six patient cohort demonstrated a 1-, 3-, and 5-year post-transplant OS of 100%, 83.3%, and 83.3%, and an RFS of 50%, respectively [71]. In a 2022 follow-up by the same group, 1-, 3-, and 5-year OS was 100%, 71%, and 57% for 18 patients receiving LT, with an RFS of 72% and 52% at 1 and 3 years [72]. An updated cohort of 26 patients showed similar outcomes, with an OS of 96% at one year and 82.7% at three years, and an RFS of 70.8% and 56.3% at one and three years, respectively [73]. In Norway, the prospective single-arm TESLA trial is also ongoing to evaluate LT for locally advanced liver-confined non-resectable iCCA that responds to neoadjuvant therapy. Similar to the Methodist study, patients receive gemcitabine-based chemotherapy for 6 months prior to LT. One chemotherapy non-responder also received Y90 SIRT. Preliminary results were recently published, demonstrating an overall transplant rate of 83% (5/6 patients) with 100% OS and 60% RFS with a median follow-up of 15 months [74]. Together, these studies suggest that LT may be considered in select patients with locally advanced disease, and the response to therapy, rather than tumor volume, could identify patients for transplant.
Given improved responses to therapy offered by alternative chemotherapy regimens, including immunotherapy and targeted therapy, there is increased interest in incorporating these regimens as neoadjuvant therapy in LT. While there is limited published data regarding these treatments prior to LT, two ongoing clinical trials (NCT06140134 [Rutgers, NJ, USA] and NCT06862934 [Milan, Italy]) include patients receiving immunotherapy and/or targeted therapies in neoadjuvant regimens. The viability of such approaches is to be determined. Only a single LT case has been reported for a patient receiving pre-LT immunotherapy. Following tumor response to gemcitabine/cisplatin/durvalumab, the patient received living donor LT with an OS and RFS of >6 months [75]. Targeted therapy with pemigatinib has also been reported in the neoadjuvant LT setting, in a patient who progressed on gemcitabine/cisplatin. After 6 months of metabolic and radiographic response to pemigatinib, the patient received LDLT with no recurrence after 1 year [76].
Incorporation of local therapies into pre-LT transplant regimens is also under investigation. In a retrospective analysis of 30 iCCA patients undergoing LT at UCLA over 30 years, the authors reported that four patients receiving combined systemic and liver-directed therapy had a 100% OS and RFS at 5 years [77]. Another intent-to-treat analysis evaluating Gem/Cis followed by TARE reported that five-year OS was 100% for the 4/17 who completed the protocol and received transplant, compared with 0% for the 13 patients who did not receive transplant [78]. Finally, a recent case report described a patient with multifocal advanced iCCA who received TARE, gemcitabine/cisplatin, and FOLFOX (folinic acid, fluorouracil, and oxaliplatin) prior to LT. Despite having a large multifocal tumor with lymphovascular involvement, the patient had a sustained treatment response prior to LT and remained recurrence-free 16 months post-transplant [79]. While these case series are limited, combined liver-directed and systemic therapy may optimize LT outcomes for locally advanced iCCA, and improved systemic options may further impact survival. Outcomes for studies evaluating LT for iCCA are summarized in Table 3. Ongoing clinical trials evaluating LT for iCCA are listed in Table 4.
Based on expanding evidence supporting the benefit of LT for iCCA, policy recommendations have recently been updated. In 2023, the AASLD updated its recommendation from “not recommended” to “consideration under research protocols” [87]. The European Association for the Study of the Liver (EASL) and the International Liver Cancer Association (ILCA) similarly recommend LT considerations for early-stage iCCA ≤3 cm [88]. Until recently, the United Network of Organ Sharing (UNOS) granted MELD exception (median MELD at transplant -3, MMaT) for pCCA but not iCCA; however, in June 2024, the OPTN introduced an updated guidance document allowing MELD exception for select iCCA. The current policy, which became active in February 2025, grants exception to patients with underlying cirrhosis who have a solitary unresectable iCCA lesion ≤3 cm and who have demonstrated tumor response or stability for >6 months [89]. While this policy will improve transplant access to patients with iCCA, the strict selection criteria will likely restrict benefit; future policy modifications allowing transplants with greater disease burden may be warranted as evidence increases. A timeline summarizing how indications have evolved for LT in iCCA is summarized in Figure 1.
Careful patient selection is critical in optimizing outcomes following LT for iCCA patients. Disease stability and response to neoadjuvant treatment have been used as surrogate indicators of favorable tumor biology [71,78]. Favorable survival for patients with significant disease burden in case series from Houston Methodist and MD Anderson suggests that tumor biology, rather than size, might indicate patients who would benefit from transplant [71]. These complex factors include genetic and epigenetic alterations (e.g., chromatin instability, DNA repair defects, loss of tumor suppressor genes, oncogene activation, aneuploidy, histone modifications, and DNA methylation), the tumor microenvironment, infiltration and activation of immune cells, cellular differentiation, tumor metabolic alterations, and genomic heterogenicity, hormonal and growth factor signaling, and immune evasion techniques by the tumor. Biologic factors are difficult to ascertain radiographically or on biopsy, leaving clinical response to therapy as the current mainstay of assessment of biologic favorability. In the future, genomic profiling, biomarker identification, circulating tumor DNA, and multiplex transcriptional and proteomic assessments may aid clinical decisions, but current data do not yet support implementation.
The role of liver transplantation for iCCA patients is a subject of ongoing investigation, but emerging data indicate that it can provide a sustained survival benefit to select patients. Our center’s opinion is that current data support expanded implementation of LT for iCCA for unresectable patients with no extrahepatic disease and disease stability for at least 6 months. The decision to offer LT needs to balance ethical issues such as organ utilization, organ availability, and predicted outcomes. In the United States, organ demand outpaces available livers for transplant, which limits the availability of deceased donors to candidates outside of the MELD exception guideline. In some European countries, deceased donor organ availability exceeds demand. We may see more aggressive expansion of iCCA transplant protocols in these locations. Given the impressive outcomes recently noted for more locally advanced iCCA, it is likely that the current UNOS selection criteria are overly restrictive. Due to the rarity of iCCA, multi-institutional or multinational prospective clinical trials would be necessary to definitively determine optimal criteria for LT. Such trials are difficult due to funding limitations, differences in center practices, and logistics. As such, an international registry to collect granular data regarding patient outcomes, radiographic and pathologic characteristics, and treatments will be necessary to obtain to more definitely determine criteria.
One alternative to expanded organ availability in iCCA is the use of living donor liver transplantation (LDLT). LDLT has been widely adopted for HCC, and is not restricted to UNOS exception criteria. As a result, several centers preferentially use LDLT for oncologic indications, such as pCCA, iCCA, and colorectal liver metastases. LDLT also allows better control of transplant timing relative to therapy, which benefits patients receiving immunotherapy or radiation, may reduce waitlist drop-out due to limited organ availability, and reduces graft ischemia-reperfusion injury. Given the risk to the healthy living donor, some feel that use of a living donor for an indication outside of current guidance is not warranted [90].
Currently, patient candidacy for transplant for iCCA is based on center-specific criteria regarding size, prior treatment, and disease stability. The rare nature of the disease and strict selection criteria may limit qualifying candidates and delay data collection regarding optimal benefit. Often, patients are referred to specialized centers only after failing multiple rounds of therapy, limiting candidacy for aggressive approaches, such as downstaging for resection or liver transplant. Early referral should be prioritized and, given the complex and evolving nature of this disease, specialized multidisciplinary patient assessment and management should be preferred.

7. Conclusions

The treatment landscape for unresectable locally advanced intrahepatic cholangiocarcinoma continues to evolve, driven by research in systemic and targeted therapies, locoregional treatments, and liver transplantation. Gemcitabine and cisplatin chemotherapy remain the standard first-line therapy for unresectable disease, but emerging immunotherapies and targeted therapies offer the potential for improved survival outcomes. Locoregional treatments, including radiation, TACE, TARE, and hepatic artery infusion, provide options for unresectable patients in select cases, and these treatments may expand access to definitive curative treatments such as surgical resection and liver transplant. A multidisciplinary approach and identifying treatment options with next-generation sequencing is vital for optimizing outcomes. Future research into targeted therapies and transplantation offers hope for improving survival and quality of life in patients with this aggressive malignancy.

Author Contributions

Conceptualization, S.L. and K.E.L.; methodology, S.L. and K.E.L.; investigation, S.L. and K.E.L.; supervision: K.E.L. and J.V.G.; writing—original draft preparation, S.L. and K.E.L.; writing—review and editing, S.L., K.E.L. and J.V.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding for this research. K.E. Lunsford and J.V. Guarrera have unrelated NIH grant funding through R21AI180739 and R01DK137222. K.E. Lunsford has additional unrelated NIH grant funding through R44HL172564.

Conflicts of Interest

The authors have no directly related conflicts of interest to report. J.V. Guarrera has received unrelated travel and research support from Organ Recovery Systems.

References

  1. Razumilava, N.; Gores, G.J. Cholangiocarcinoma. Lancet 2014, 383, 2168–2179. [Google Scholar] [CrossRef]
  2. Rodrigues, P.M.; Olaizola, P.; Paiva, N.A.; Olaizola, I.; Agirre-Lizaso, A.; Landa, A.; Bujanda, L.; Perugorria, M.J.; Banales, J.M. Pathogenesis of Cholangiocarcinoma. Annu. Rev. Pathol. 2021, 16, 433–463. [Google Scholar] [CrossRef] [PubMed]
  3. Panayotova, G.; Guerra, J.; Guarrera, J.V.; Lunsford, K.E. The Role of Surgical Resection and Liver Transplantation for the Treatment of Intrahepatic Cholangiocarcinoma. J. Clin. Med. 2021, 10, 2428. [Google Scholar] [CrossRef] [PubMed]
  4. Shin, D.W.; Moon, S.H.; Kim, J.H. Diagnosis of Cholangiocarcinoma. Diagnostics 2023, 13, 233. [Google Scholar] [CrossRef]
  5. Connor, A.A.; Kodali, S.; Abdelrahim, M.; Javle, M.M.; Brombosz, E.W.; Ghobrial, R.M. Intrahepatic cholangiocarcinoma: The role of liver transplantation, adjunctive treatments, and prognostic biomarkers. Front. Oncol. 2022, 12, 996710. [Google Scholar] [CrossRef] [PubMed]
  6. Wang, Y.; Alsaraf, Y.; Bandaru, S.S.; Lyons, S.; Reap, L.; Ngo, T.; Yu, Z.; Yu, Q. Epidemiology, survival and new treatment modalities for intrahepatic cholangiocarcinoma. J. Gastrointest. Oncol. 2024, 15, 1777–1788. [Google Scholar] [CrossRef]
  7. Banales, J.M.; Marin, J.J.G.; Lamarca, A.; Rodrigues, P.M.; Khan, S.A.; Roberts, L.R.; Cardinale, V.; Carpino, G.; Andersen, J.B.; Braconi, C.; et al. Cholangiocarcinoma 2020: The next horizon in mechanisms and management. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 557–588. [Google Scholar] [CrossRef]
  8. Cerrito, L.; Ainora, M.E.; Borriello, R.; Piccirilli, G.; Garcovich, M.; Riccardi, L.; Pompili, M.; Gasbarrini, A.; Zocco, M.A. Contrast-Enhanced Imaging in the Management of Intrahepatic Cholangiocarcinoma: State of Art and Future Perspectives. Cancers 2023, 15, 3393. [Google Scholar] [CrossRef]
  9. Weber, S.M.; Ribero, D.; O’Reilly, E.M.; Kokudo, N.; Miyazaki, M.; Pawlik, T.M. Intrahepatic cholangiocarcinoma: Expert consensus statement. HPB 2015, 17, 669–680. [Google Scholar] [CrossRef]
  10. Orcutt, S.T.; Anaya, D.A. Liver Resection and Surgical Strategies for Management of Primary Liver Cancer. Cancer Control 2018, 25, 1073274817744621. [Google Scholar] [CrossRef]
  11. Mavros, M.N.; Economopoulos, K.P.; Alexiou, V.G.; Pawlik, T.M. Treatment and Prognosis for Patients with Intrahepatic Cholangiocarcinoma: Systematic Review and Meta-analysis. JAMA Surg. 2014, 149, 565–574. [Google Scholar] [CrossRef] [PubMed]
  12. de Jong, M.C.; Nathan, H.; Sotiropoulos, G.C.; Paul, A.; Alexandrescu, S.; Marques, H.; Pulitano, C.; Barroso, E.; Clary, B.M.; Aldrighetti, L.; et al. Intrahepatic cholangiocarcinoma: An international multi-institutional analysis of prognostic factors and lymph node assessment. J. Clin. Oncol. 2011, 29, 3140–3145. [Google Scholar] [CrossRef] [PubMed]
  13. Li, T.; Qin, L.X.; Zhou, J.; Sun, H.C.; Qiu, S.J.; Ye, Q.H.; Wang, L.; Tang, Z.Y.; Fan, J. Staging, prognostic factors and adjuvant therapy of intrahepatic cholangiocarcinoma after curative resection. Liver Int. 2014, 34, 953–960. [Google Scholar] [CrossRef] [PubMed]
  14. Ribero, D.; Pinna, A.D.; Guglielmi, A.; Ponti, A.; Nuzzo, G.; Giulini, S.M.; Aldrighetti, L.; Calise, F.; Gerunda, G.E.; Tomatis, M.; et al. Surgical Approach for Long-term Survival of Patients with Intrahepatic Cholangiocarcinoma: A Multi-institutional Analysis of 434 Patients. Arch. Surg. 2012, 147, 1107–1113. [Google Scholar] [CrossRef]
  15. Farges, O.; Fuks, D.; Boleslawski, E.; Le Treut, Y.P.; Castaing, D.; Laurent, A.; Ducerf, C.; Rivoire, M.; Bachellier, P.; Chiche, L.; et al. Influence of surgical margins on outcome in patients with intrahepatic cholangiocarcinoma: A multicenter study by the AFC-IHCC-2009 study group. Ann. Surg. 2011, 254, 824–829, discussion 830. [Google Scholar] [CrossRef]
  16. Primrose, J.N.; Fox, R.P.; Palmer, D.H.; Malik, H.Z.; Prasad, R.; Mirza, D.; Anthony, A.; Corrie, P.; Falk, S.; Finch-Jones, M.; et al. Capecitabine compared with observation in resected biliary tract cancer (BILCAP): A randomised, controlled, multicentre, phase 3 study. Lancet Oncol. 2019, 20, 663–673. [Google Scholar] [CrossRef]
  17. Edeline, J.; Benabdelghani, M.; Bertaut, A.; Watelet, J.; Hammel, P.; Joly, J.P.; Boudjema, K.; Fartoux, L.; Bouhier-Leporrier, K.; Jouve, J.L.; 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, 658–667. [Google Scholar] [CrossRef]
  18. Neoptolemos, J.P.; Moore, M.J.; Cox, T.F.; Valle, J.W.; Palmer, D.H.; McDonald, A.C.; Carter, R.; Tebbutt, N.C.; Dervenis, C.; Smith, D.; et al. Effect of adjuvant chemotherapy with fluorouracil plus folinic acid or gemcitabine vs observation on survival in patients with resected periampullary adenocarcinoma: The ESPAC-3 periampullary cancer randomized trial. JAMA 2012, 308, 147–156. [Google Scholar] [CrossRef]
  19. Shroff, R.T.; Kennedy, E.B.; Bachini, M.; Bekaii-Saab, T.; Crane, C.; Edeline, J.; El-Khoueiry, A.; Feng, M.; Katz, M.H.G.; Primrose, J.; et al. Adjuvant Therapy for Resected Biliary Tract Cancer: ASCO Clinical Practice Guideline. J. Clin. Oncol. 2019, 37, 1015–1027. [Google Scholar] [CrossRef]
  20. Valle, J.; Wasan, H.; Palmer, D.H.; Cunningham, D.; Anthoney, A.; Maraveyas, A.; Madhusudan, S.; Iveson, T.; Hughes, S.; Pereira, S.P.; et al. Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. N. Engl. J. Med. 2010, 362, 1273–1281. [Google Scholar] [CrossRef]
  21. Oh, D.Y.; He, A.R.; Qin, S.; Chen, L.T.; Okusaka, T.; Vogel, A.; Kim, J.W.; Suksombooncharoen, T.; Lee, M.A.; Kitano, M.; et al. Durvalumab plus Gemcitabine and Cisplatin in Advanced Biliary Tract Cancer. NEJM Evid. 2022, 1, EVIDoa2200015. [Google Scholar] [CrossRef] [PubMed]
  22. Kelley, R.K.; Ueno, M.; Yoo, C.; Finn, R.S.; Furuse, J.; Ren, Z.; Yau, T.; Klümpen, H.J.; Chan, S.L.; Ozaka, M.; 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, 1853–1865. [Google Scholar] [CrossRef] [PubMed]
  23. Shroff, R.T.; Javle, M.M.; Xiao, L.; Kaseb, A.O.; Varadhachary, G.R.; Wolff, R.A.; Raghav, K.P.S.; Iwasaki, M.; Masci, P.; Ramanathan, R.K.; et al. Gemcitabine, Cisplatin, and nab-Paclitaxel for the Treatment of Advanced Biliary Tract Cancers: A Phase 2 Clinical Trial. JAMA Oncol. 2019, 5, 824–830. [Google Scholar] [CrossRef]
  24. Shroff, R.T.; King, G.; Colby, S.; Scott, A.J.; Borad, M.J.; Goff, L.; Matin, K.; Mahipal, A.; Kalyan, A.; Javle, M.M.; et al. SWOG S1815: 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. 2025, 43, 536–544. [Google Scholar] [CrossRef]
  25. Yadav, S.; Xie, H.; Bin-Riaz, I.; Sharma, P.; Durani, U.; Goyal, G.; Borah, B.; Borad, M.J.; Smoot, R.L.; Roberts, L.R.; et al. Neoadjuvant vs. adjuvant chemotherapy for cholangiocarcinoma: A propensity score matched analysis. Eur. J. Surg. Oncol. 2019, 45, 1432–1438. [Google Scholar] [CrossRef]
  26. Rizzo, A.; Brandi, G. Neoadjuvant therapy for cholangiocarcinoma: A comprehensive literature review. Cancer Treat. Res. Commun. 2021, 27, 100354. [Google Scholar] [CrossRef] [PubMed]
  27. Le Roy, B.; Gelli, M.; Pittau, G.; Allard, M.A.; Pereira, B.; Serji, B.; Vibert, E.; Castaing, D.; Adam, R.; Cherqui, D.; et al. Neoadjuvant chemotherapy for initially unresectable intrahepatic cholangiocarcinoma. Br. J. Surg. 2018, 105, 839–847. [Google Scholar] [CrossRef]
  28. Maithel, S.K.; Keilson, J.M.; Cao, H.S.T.; Rupji, M.; Mahipal, A.; Lin, B.S.; Javle, M.M.; Cleary, S.P.; Akce, M.; Switchenko, J.M.; et al. NEO-GAP: A Single-Arm, Phase II Feasibility Trial of Neoadjuvant Gemcitabine, Cisplatin, and Nab-Paclitaxel for Resectable, High-Risk Intrahepatic Cholangiocarcinoma. Ann. Surg. Oncol. 2023, 30, 6558–6566. [Google Scholar] [CrossRef]
  29. U.S. National Library of Medicine. Gemcitabine, Cisplatin, and Nab-Paclitaxel Before Surgery in Patients with High-Risk Liver Bile Duct Cancer. Available online: https://clinicaltrials.gov/study/NCT03579771 (accessed on 6 May 2025).
  30. U.S. National Library of Medicine. Durvalumab with Gemcitabine and Cisplatin for the Treatment of High-Risk Resectable Liver Cancer Before Surgery. Available online: https://clinicaltrials.gov/study/NCT06050252 (accessed on 2 May 2025).
  31. U.S. National Library of Medicine. A Single-Arm Study of Pembrolizumab with Gemcitabine and Cisplatin as Perioperative Therapy for Potentially Resectable Intrahepatic Cholangiocarcinoma. Available online: https://clinicaltrials.gov/study/NCT05967182 (accessed on 2 May 2025).
  32. U.S. National Library of Medicine. Gemcitabine/Cisplatin/Nab-Paclitaxel and Rilvegostomig in Resectable iCCA (NEOLANGIO). Available online: https://clinicaltrials.gov/study/NCT06569225 (accessed on 2 May 2025).
  33. Lowery, M.A.; Ptashkin, R.; Jordan, E.; Berger, M.F.; Zehir, A.; Capanu, M.; Kemeny, N.E.; O’Reilly, E.M.; El-Dika, I.; Jarnagin, W.R.; et al. Comprehensive Molecular Profiling of Intrahepatic and Extrahepatic Cholangiocarcinomas: Potential Targets for Intervention. Clin. Cancer Res. 2018, 24, 4154–4161. [Google Scholar] [CrossRef]
  34. Abou-Alfa, G.K.; Sahai, V.; Hollebecque, A.; Vaccaro, G.; Melisi, D.; Al-Rajabi, R.; Paulson, A.S.; Borad, M.J.; Gallinson, D.; Murphy, A.G.; et al. Pemigatinib for previously treated, locally advanced or metastatic cholangiocarcinoma: A multicentre, open-label, phase 2 study. Lancet Oncol. 2020, 21, 671–684. [Google Scholar] [CrossRef]
  35. Goyal, L.; Meric-Bernstam, F.; Hollebecque, A.; Valle, J.W.; Morizane, C.; Karasic, T.B.; Abrams, T.A.; Furuse, J.; Kelley, R.K.; Cassier, P.A.; et al. Futibatinib for FGFR2-Rearranged Intrahepatic Cholangiocarcinoma. N. Engl. J. Med. 2023, 388, 228–239. [Google Scholar] [CrossRef] [PubMed]
  36. Zhu, A.X.; Macarulla, T.; Javle, M.M.; Kelley, R.K.; Lubner, S.J.; Adeva, J.; Cleary, J.M.; Catenacci, D.V.T.; Borad, M.J.; Bridgewater, J.A.; 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, 1669–1677. [Google Scholar] [CrossRef] [PubMed]
  37. Hong, T.S.; Wo, J.Y.; Yeap, B.Y.; Ben-Josef, E.; McDonnell, E.I.; Blaszkowsky, L.S.; Kwak, E.L.; Allen, J.N.; Clark, J.W.; Goyal, L.; et al. Multi-Institutional Phase II Study of High-Dose Hypofractionated Proton Beam Therapy in Patients with Localized, Unresectable Hepatocellular Carcinoma and Intrahepatic Cholangiocarcinoma. J. Clin. Oncol. 2016, 34, 460–468. [Google Scholar] [CrossRef]
  38. Tao, R.; Krishnan, S.; Bhosale, P.R.; Javle, M.M.; Aloia, T.A.; Shroff, R.T.; Kaseb, A.O.; Bishop, A.J.; Swanick, C.W.; Koay, E.J.; 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, 219–226. [Google Scholar] [CrossRef] [PubMed]
  39. Yang, A.H.; Urrunaga, N.H.; Siddiqui, O.; Wu, A.; Schliep, M.; Mossahebi, S.; Shetty, K.; Regine, W.F.; Molitoris, J.K.; Lominadze, Z. Proton beam stereotactic body radiotherapy and hypofractionated therapy with pencil beam scanning is safe and effective for advanced hepatocellular carcinoma and intrahepatic cholangiocarcinoma: A single center experience. J. Radiosurg. SBRT 2023, 9, 43–52. [Google Scholar]
  40. Bourien, H.; Pircher, C.C.; Guiu, B.; Lamarca, A.; Valle, J.W.; Niger, M.; Edeline, J. Locoregional Treatment in Intrahepatic Cholangiocarcinoma: Which Treatment for Which Patient? Cancers 2023, 15, 4217. [Google Scholar] [CrossRef] [PubMed]
  41. Zhou, T.Y.; Zhou, G.H.; Zhang, Y.L.; Nie, C.H.; Zhu, T.Y.; Wang, H.L.; Chen, S.Q.; Wang, B.Q.; Yu, Z.N.; Wu, L.M.; et al. Drug-eluting beads transarterial chemoembolization with CalliSpheres microspheres for treatment of unresectable intrahepatic cholangiocarcinoma. J. Cancer 2020, 11, 4534–4541. [Google Scholar] [CrossRef]
  42. Park, S.Y.; Kim, J.H.; Yoon, H.J.; Lee, I.S.; Yoon, H.K.; Kim, K.P. Transarterial chemoembolization versus supportive therapy in the palliative treatment of unresectable intrahepatic cholangiocarcinoma. Clin. Radiol. 2011, 66, 322–328. [Google Scholar] [CrossRef]
  43. Vogl, T.J.; Naguib, N.N.; Nour-Eldin, N.E.; Bechstein, W.O.; Zeuzem, S.; Trojan, J.; Gruber-Rouh, T. Transarterial chemoembolization in the treatment of patients with unresectable cholangiocarcinoma: Results and prognostic factors governing treatment success. Int. J. Cancer 2012, 131, 733–740. [Google Scholar] [CrossRef]
  44. Kiefer, M.V.; Albert, M.; McNally, M.; Robertson, M.; Sun, W.; Fraker, D.; Olthoff, K.; Christians, K.; Pappas, S.; Rilling, W.; et al. Chemoembolization of intrahepatic cholangiocarcinoma with cisplatinum, doxorubicin, mitomycin C, ethiodol, and polyvinyl alcohol: A 2-center study. Cancer 2011, 117, 1498–1505. [Google Scholar] [CrossRef]
  45. Martin, R.C.G., 2nd; Simo, K.A.; Hansen, P.; Rocha, F.; Philips, P.; McMasters, K.M.; Tatum, C.M.; Kelly, L.R.; Driscoll, M.; Sharma, V.R.; et al. Drug-Eluting Bead, Irinotecan Therapy of Unresectable Intrahepatic Cholangiocarcinoma (DELTIC) with Concomitant Systemic Gemcitabine and Cisplatin. Ann. Surg. Oncol. 2022, 29, 5462–5473. [Google Scholar] [CrossRef] [PubMed]
  46. Edeline, J.; Touchefeu, Y.; Guiu, B.; Farge, O.; Tougeron, D.; Baumgaertner, I.; Ayav, A.; Campillo-Gimenez, B.; Beuzit, L.; Pracht, M.; et al. Radioembolization Plus Chemotherapy for First-line Treatment of Locally Advanced Intrahepatic Cholangiocarcinoma: A Phase 2 Clinical Trial. JAMA Oncol. 2020, 6, 51–59. [Google Scholar] [CrossRef] [PubMed]
  47. Schartz, D.A.; Porter, M.; Schartz, E.; Kallas, J.; Gupta, A.; Butani, D.; Cantos, A. Transarterial Yttrium-90 Radioembolization for Unresectable Intrahepatic Cholangiocarcinoma: A Systematic Review and Meta-Analysis. J. Vasc. Interv. Radiol. 2022, 33, 679–686. [Google Scholar] [CrossRef]
  48. Chan, S.L.; Chotipanich, C.; Choo, S.P.; Kwang, S.W.; Mo, F.; Worakitsitisatorn, A.; Tai, D.; Sundar, R.; Ng, D.C.E.; Loke, K.S.H.; et al. Selective Internal Radiation Therapy with Yttrium-90 Resin Microspheres Followed by Gemcitabine plus Cisplatin for Unresectable Intrahepatic Cholangiocarcinoma: A Phase 2 Single-Arm Multicenter Clinical Trial. Liver Cancer 2022, 11, 451–459. [Google Scholar] [CrossRef]
  49. Moris, D.; Palta, M.; Kim, C.; Allen, P.J.; Morse, M.A.; Lidsky, M.E. Advances in the treatment of intrahepatic cholangiocarcinoma: An overview of the current and future therapeutic landscape for clinicians. CA Cancer J. Clin. 2023, 73, 198–222. [Google Scholar] [CrossRef] [PubMed]
  50. Massani, M.; Bonariol, L.; Stecca, T. Hepatic Arterial Infusion Chemotherapy for Unresectable Intrahepatic Cholangiocarcinoma, a Comprehensive Review. J. Clin. Med. 2021, 10, 2552. [Google Scholar] [CrossRef]
  51. Boehm, L.M.; Jayakrishnan, T.T.; Miura, J.T.; Zacharias, A.J.; Johnston, F.M.; Turaga, K.K.; Gamblin, T.C. Comparative effectiveness of hepatic artery based therapies for unresectable intrahepatic cholangiocarcinoma. J. Surg. Oncol. 2015, 111, 213–220. [Google Scholar] [CrossRef]
  52. Konstantinidis, I.T.; Groot Koerkamp, B.; Do, R.K.; Gönen, M.; Fong, Y.; Allen, P.J.; D’Angelica, M.I.; Kingham, T.P.; DeMatteo, R.P.; Klimstra, D.S.; 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, 758–765. [Google Scholar] [CrossRef]
  53. Cercek, A.; Boerner, T.; Tan, B.R.; Chou, J.F.; Gönen, M.; Boucher, T.M.; Hauser, H.F.; Do, R.K.G.; Lowery, M.A.; Harding, J.J.; 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, 60–67. [Google Scholar] [CrossRef]
  54. Zhao, R.; Zhou, J.; Miao, Z.; Xiong, X.; Wei, W.; Li, S.; Guo, R. Efficacy and safety of lenvatinib plus durvalumab combined with hepatic arterial infusion chemotherapy for unresectable intrahepatic cholangiocarcinoma. Front. Immunol. 2024, 15, 1397827. [Google Scholar]
  55. Mayo, S.C.; Patel, R.K.; Walker, B.S.; Eil, R.; Wong, M.; Fung, A.; Brody, J.R.; Anand, S.; Corless, C.L.; Hansen, L.; et al. A phase II trial of induction systemic mFOLFIRINOX followed by hepatic arterial infusion of floxuridine and dexamethasone given concurrently with systemic mFOLFIRI as a first-line therapy in patients with unresectable liver-dominant intrahepatic cholangiocarcinoma (HELIX-1). J. Clin. Oncol. 2024, 42, 511. [Google Scholar]
  56. Zhu, M.; Jin, M.; Zhao, X.; Shen, S.; Chen, Y.; Xiao, H.; Wei, G.; He, Q.; Li, B.; Peng, Z. Anti-PD-1 antibody in combination with radiotherapy as first-line therapy for unresectable intrahepatic cholangiocarcinoma. BMC Med. 2024, 22, 165. [Google Scholar] [CrossRef]
  57. Victory, J.H.; Smith, E.C.; Ryan, C.E.; Lambdin, J.; Sarvestani, A.L.; Friedman, L.R.; Eade, A.V.; Larrain, C.; Pu, T.; Luberice, K.; et al. Hepatic artery infusion pump (HAIP) therapy in combination with targeted delivery of IL-12 for patients with metastatic colorectal cancer or intrahepatic cholangiocarcinoma: A phase II trial protocol. J. Gastrointest. Oncol. 2024, 15, 1348–1354. [Google Scholar] [CrossRef] [PubMed]
  58. Hendricks-Wenger, A.; Weber, P.; Simon, A.; Saunier, S.; Coutermarsh-Ott, S.; Grider, D.; Vidal-Jove, J.; Allen, I.C.; Luyimbazi, D.; Vlaisavljevich, E. Histotripsy for the Treatment of Cholangiocarcinoma Liver Tumors: In Vivo Feasibility and Ex Vivo Dosimetry Study. IEEE Trans. Ultrason. Ferroelectr. Freq. Control 2021, 68, 2953–2964. [Google Scholar] [CrossRef]
  59. Sun, D.; Lv, G.; Dong, J. Liver Transplantation for Intrahepatic Cholangiocarcinoma: What Are New Insights and What Should We Follow? Front. Oncol. 2021, 11, 841694. [Google Scholar] [CrossRef]
  60. Kodali, S.; Connor, A.A.; Thabet, S.; Brombosz, E.W.; Ghobrial, R.M. Liver transplantation as an alternative for the treatment of intrahepatic cholangiocarcinoma: Past, present, and future directions. Hepatobiliary Pancreat. Dis. Int. 2024, 23, 129–138. [Google Scholar] [CrossRef] [PubMed]
  61. Penn, I. Hepatic transplantation for primary and metastatic cancers of the liver. Surgery 1991, 110, 726–734, discussion 734–735. [Google Scholar]
  62. Borakati, A.; Froghi, F.; Bhogal, R.H.; Mavroeidis, V.K. Liver transplantation in the management of cholangiocarcinoma: Evolution and contemporary advances. World J. Gastroenterol. 2023, 29, 1969–1981. [Google Scholar] [CrossRef]
  63. Sapisochin, G.; de Lope, C.R.; Gastaca, M.; de Urbina, J.O.; Suarez, M.A.; Santoyo, J.; Castroagudín, J.F.; Varo, E.; López-Andujar, R.; Palacios, F.; et al. “Very early” intrahepatic cholangiocarcinoma in cirrhotic patients: Should liver transplantation be reconsidered in these patients? Am. J. Transplant. 2014, 14, 660–667. [Google Scholar] [CrossRef]
  64. Sapisochin, G.; Facciuto, M.; Rubbia-Brandt, L.; Marti, J.; Mehta, N.; Yao, F.Y.; Vibert, E.; Cherqui, D.; Grant, D.R.; Hernandez-Alejandro, R.; et al. Liver transplantation for “very early” intrahepatic cholangiocarcinoma: International retrospective study supporting a prospective assessment. Hepatology 2016, 64, 1178–1188. [Google Scholar] [CrossRef]
  65. De Martin, E.; Rayar, M.; Golse, N.; Dupeux, M.; Gelli, M.; Gnemmi, V.; Allard, M.A.; Cherqui, D.; Sa Cunha, A.; Adam, R.; et al. Analysis of Liver Resection Versus Liver Transplantation on Outcome of Small Intrahepatic Cholangiocarcinoma and Combined Hepatocellular-Cholangiocarcinoma in the Setting of Cirrhosis. Liver Transpl. 2020, 26, 785–798. [Google Scholar] [CrossRef] [PubMed]
  66. Sempoux, C.; Jibara, G.; Ward, S.C.; Fan, C.; Qin, L.; Roayaie, S.; Fiel, M.I.; Schwartz, M.; Thung, S.N. Intrahepatic cholangiocarcinoma: New insights in pathology. Semin. Liver Dis. 2011, 31, 49–60. [Google Scholar] [CrossRef] [PubMed]
  67. Chu, K.J.; Lu, C.D.; Dong, H.; Fu, X.H.; Zhang, H.W.; Yao, X.P. Hepatitis B virus-related combined hepatocellular-cholangiocarcinoma: Clinicopathological and prognostic analysis of 390 cases. Eur. J. Gastroenterol. Hepatol. 2014, 26, 192–199. [Google Scholar] [CrossRef] [PubMed]
  68. Lunsford, K.E.; Court, C.; Lee, Y.S.; Lu, D.S.; Naini, B.V.; Harlander-Locke, M.P.; Busuttil, R.W.; Agopian, V.G. Propensity-Matched Analysis of Patients with Mixed Hepatocellular-Cholangiocarcinoma and Hepatocellular Carcinoma Undergoing Liver Transplantation. Liver Transpl. 2018, 24, 1384–1397. [Google Scholar] [CrossRef]
  69. Dageforde, L.A.; Vachharajani, N.; Tabrizian, P.; Agopian, V.; Halazun, K.; Maynard, E.; Croome, K.; Nagorney, D.; Hong, J.C.; Lee, D.; et al. Multi-Center Analysis of Liver Transplantation for Combined Hepatocellular Carcinoma-Cholangiocarcinoma Liver Tumors. J. Am. Coll. Surg. 2021, 232, 361–371. [Google Scholar] [CrossRef]
  70. Hong, J.C.; Jones, C.M.; Duffy, J.P.; Petrowsky, H.; Farmer, D.G.; French, S.; Finn, R.; Durazo, F.A.; Saab, S.; Tong, M.J.; et al. Comparative analysis of resection and liver transplantation for intrahepatic and hilar cholangiocarcinoma: A 24-year experience in a single center. Arch. Surg. 2011, 146, 683–689. [Google Scholar] [CrossRef]
  71. Lunsford, K.E.; Javle, M.; Heyne, K.; Shroff, R.T.; Abdel-Wahab, R.; Gupta, N.; Mobley, C.M.; Saharia, A.; Victor, D.W.; Nguyen, D.T.; et al. Liver transplantation for locally advanced intrahepatic cholangiocarcinoma treated with neoadjuvant therapy: A prospective case-series. Lancet Gastroenterol. Hepatol. 2018, 3, 337–348. [Google Scholar] [CrossRef]
  72. McMillan, R.R.; Javle, M.; Kodali, S.; Saharia, A.; Mobley, C.; Heyne, K.; Hobeika, M.J.; Lunsford, K.E.; Victor, D.W., 3rd; Shetty, A.; et al. Survival following liver transplantation for locally advanced, unresectable intrahepatic cholangiocarcinoma. Am. J. Transplant. 2022, 22, 823–832. [Google Scholar] [CrossRef]
  73. Semaan, S.; Connor, A.A.; Saharia, A.; Kodali, S.; Elaileh, A.; Patel, K.; Soliman, N.; Basra, T.; Victor, D.W., 3rd; Simon, C.J.; et al. Transplantation for Peri-Hilar and Intrahepatic Cholangiocarcinoma with mTOR Immunosuppression. Transplant. Proc. 2025, 57, 255–263. [Google Scholar] [CrossRef]
  74. Yaqub, S.; Busund, S.; Smedman, T.M.; Syversveen, T.; Khan, A.; Solheim, J.M.; Folseraas, T.; Wiencke, K.; Lassen, K.; Dueland, S.; et al. Liver transplantation for locally advanced non-resectable intrahepatic cholangiocarcinoma treated with neoadjuvant therapy: Early results from the TESLA trial. Br. J. Surg. 2025, 112, znaf054. [Google Scholar] [CrossRef]
  75. Fernandes, E.S.M.; Mello, F.P.T.; Andrade, R.O.; Girão, C.L.; Cesar, C.; Pimentel, L.S.; Coelho, H.S.M.; Basto, S.T.; Siqueira, M.; Brito, A.; et al. Living donor liver transplant for intrahepatic cholangiocarcinoma. An initial brazilian experience. Arq. Bras. Cir. Dig. 2024, 37, e1839. [Google Scholar] [CrossRef] [PubMed]
  76. Byrne, M.M.; Dunne, R.F.; Melaragno, J.I.; Chávez-Villa, M.; Hezel, A.; Liao, X.; Ertreo, M.; Al-Judaibi, B.; Orloff, M.; Hernandez-Alejandro, R.; et al. Neoadjuvant pemigatinib as a bridge to living donor liver transplantation for intrahepatic cholangiocarcinoma with FGFR2 gene rearrangement. Am. J. Transplant. 2025, 25, 623–627. [Google Scholar] [CrossRef] [PubMed]
  77. Ito, T.; Butler, J.R.; Noguchi, D.; Ha, M.; Aziz, A.; Agopian, V.G.; DiNorcia, J., 3rd; Yersiz, H.; Farmer, D.G.; Busuttil, R.W.; et al. A 3-Decade, Single-Center Experience of Liver Transplantation for Cholangiocarcinoma: Impact of Era, Tumor Size, Location, and Neoadjuvant Therapy. Liver Transpl. 2022, 28, 386–396. [Google Scholar] [CrossRef] [PubMed]
  78. Maspero, M.; Sposito, C.; Bongini, M.A.; Cascella, T.; Flores, M.; Maccauro, M.; Chiesa, C.; Niger, M.; Pietrantonio, F.; Leoncini, G.; et al. Liver Transplantation for Intrahepatic Cholangiocarcinoma After Chemotherapy and Radioembolization: An Intention-To-Treat Study. Transpl. Int. 2024, 37, 13641. [Google Scholar] [CrossRef]
  79. Teixeira, C.; Viamonte, B.; Graça, L.; Pinto Marques, H.; Rego, I.; Ribeiro, M.J. Liver Transplant After Neoadjuvant Treatment for Long-Term Survivors with Intrahepatic Cholangiocarcinoma: Does It Have a Role? Cureus 2024, 16, e75935. [Google Scholar] [CrossRef]
  80. U.S. National Library of Medicine. Liver Transplantation for Early Intrahepatic Cholangiocarcinoma (LT for iCCA). Available online: https://clinicaltrials.gov/study/NCT02878473 (accessed on 6 May 2025).
  81. U.S. National Library of Medicine. Liver Transplant for Stable, Advanced Intrahepatic Cholangiocarcinoma. Available online: https://clinicaltrials.gov/study/NCT04195503 (accessed on 6 May 2025).
  82. U.S. National Library of Medicine. Liver Transplantation for Non-Resectable Intrahepatic Cholangiocarcinoma: A Prospective Exploratory Trial (TESLA Trial). Available online: https://clinicaltrials.gov/study/NCT04556214 (accessed on 6 May 2025).
  83. U.S. National Library of Medicine. Liver Transplantation in Intrahepatic Cholangiocarcinoma. Available online: https://clinicaltrials.gov/study/NCT06140134 (accessed on 6 May 2025).
  84. U.S. National Library of Medicine. LIver Transplantation for Non-Resectable Intrahepatic CholAngiocarcinoma (LIRICA) (LIRICA). Available online: https://clinicaltrials.gov/study/NCT06098547 (accessed on 6 May 2025).
  85. U.S. National Library of Medicine. Liver Transplantation for Unresectable Intrahepatic Colangiocarcinoma After Sustained Response to Neoadjuvant Treatments (iCOLA). Available online: https://clinicaltrials.gov/study/NCT06862934 (accessed on 6 May 2025).
  86. U.S. National Library of Medicine. Living Donor Liver Transplantation for Intrahepatic Cholangiocarcinoma (LIVINCA). Available online: https://clinicaltrials.gov/study/NCT06539377 (accessed on 6 May 2025).
  87. Bowlus, C.L.; Arrivé, L.; Bergquist, A.; Deneau, M.; Forman, L.; Ilyas, S.I.; Lunsford, K.E.; Martinez, M.; Sapisochin, G.; Shroff, R.; et al. AASLD practice guidance on primary sclerosing cholangitis and cholangiocarcinoma. Hepatology 2023, 77, 659–702. [Google Scholar] [CrossRef]
  88. EASL-ILCA Clinical Practice Guidelines on the management of intrahepatic cholangiocarcinoma. J. Hepatol. 2023, 79, 181–208. [CrossRef]
  89. National Liver Review Board (NLRB). Updates Related to Transplant Oncology—Public Comment Proposal; Organ Procurement and Transplantation Network Liver & Intestinal Organ Transplantation Committee: Washington, DC, USA, 2024. [Google Scholar]
  90. Andraus, W.; Ochoa, G.; de Martino, R.B.; Pinheiro, R.S.N.; Santos, V.R.; Lopes, L.D.; Júnior, R.M.A.; Waisberg, D.R.; Santana, A.C.; Tustumi, F.; et al. The role of living donor liver transplantation in treating intrahepatic cholangiocarcinoma. Front. Oncol. 2024, 14, 1404683. [Google Scholar] [CrossRef]
Figure 1. Timeline of evolving indications for LT for iCCA [87,88,89].
Figure 1. Timeline of evolving indications for LT for iCCA [87,88,89].
Curroncol 32 00293 g001
Table 1. Prospective trials investigating neoadjuvant regimens for iCCA.
Table 1. Prospective trials investigating neoadjuvant regimens for iCCA.
ReferenceInterventionStudy StatusEndpointResults
Maithel 2023 [28]Gemcitabine/cisplatin/nab-paclitaxel + resectionMulti-institutional, phase IICompletePrimary: completion of both preoperative chemotherapy + resection
Secondary: AEs, radiologic response, RFS and OS
Primary: 30 completed preoperative chemotherapy and 22 were resected
Secondary: 90%
with disease control, 23% with partial response
Median OS 24 months
Median RFS 7.1 months
NCT03579771 [29]Gemcitabine/cisplatin/nab-paclitaxel + FGFR2 inhibitor (for patients with FGFR2 fusion or
rearrangement) +
resection
Single-arm,
phase II
Active,
notrecruiting
Primary: completion of all preoperative + operative therapy, safety
Secondary: radiological response, RFS, OS
-
NCT06050252 [30]Gemcitabine/cisplatin/durvalumab +
resection
Multi-
institutional, phase II
Actively
recruiting
Primary: completion rate of neoadjuvant treatment + resection
Secondary: major pathologic response
-
NCT05967182 [31]Gemcitabine/
cisplatin/
pembrolizumab +
resection
Single-arm, phase II trialActively
recruiting
Primary: RFS and major pathologic
response
-
NCT06569225 [32]Gemcitabine/
cisplatin/
nab-paclitaxel/
rilvegostomig +
resection
Multi-
institutional, phase II
Not yet
recruiting
Primary: major pathologic response
Secondary: radiologic objective response rate, AEs, rate of R0
-
Table 2. Comparison of OS and PFS in prospective trials for liver-directed therapies for unresectable iCCA.
Table 2. Comparison of OS and PFS in prospective trials for liver-directed therapies for unresectable iCCA.
ReferenceInterventionStudynMedian OSMedian PFS
Hong 2016 [37]High-dose
hypofractionated proton beam therapy
Phase II,
multi-institutional single-arm study
3722.5 months (95% CI 12.4–49.7)8.4 months (95% CI 5.0–15.7)
Martin 2022 [45]DEBIRI TACE + Gem/CisPhase II,
multicenter randomized study
4833.7 months (95% CI 13.5–54.5)31.9 months (95% CI 8.5–75.3)
Edeline 2020 [46]TARE + Gem/CisPhase II,
multicenter clinical trial
4122 months (95% CI 14–52)14 months (95% CI 8–17 months)
Chan 2022 [48]TARE + Gem/CisPhase II,
multicenter single-arm clinical trial
1621.6 months (95% CI 7.3–25.2)9 months (95% CI 3.2–13.1)
Cercek 2020 [53] HAI floxuridine +
systemic Gem/Ox
Phase II,
single-arm clinical trial
3825 months (95% CI, 20.60 not reached)11.7 months (1-sided 95% CI 11.1)
Table 3. Comparison of outcomes for studies evaluating LT for iCCA.
Table 3. Comparison of outcomes for studies evaluating LT for iCCA.
ReferenceStudy DesignPopulation InterventionKey Outcomes
Penn 1991 [61]RetrospectiveiCCA patientsLT2- and 5-yr OS: 30%, 17%
Recurrence: 44%
Sapisochin 2014 [63]Retrospective
multicenter
29 iCCA patients with cirrhosis (8 with “very early” iCCA ≤ 2 cm)LTIn “very early” subgroup:
Recurrence: 0%
OS 1-, 3-, 5-yr: 100%, 73%, 73%
Sapisochin 2016 [64]Retrospective
multicenter
“Very early” iCCA (≤2 cm) vs. advanced iCCA (>2 cm or multifocal) LTRecurrence at 1, 3, and 5 yrs: 7%, 18%, 18% (“very early”) vs. 30%, 47%, 61%;
OS: 93%, 84%, 65% vs. 79%, 50%, 45%
De Martin 2020 [65]Retrospective
multicenter
Cirrhotic patients with iCCA or combined HCC and cholangiocarcinoma ≤ 5 cmLT vs.
resection
5-yr RFS: 75% (LT) vs. 36% (resection)
For tumors 2–5 cm: recurrence 21% (LT) vs. 48% (resection); RFS 74% vs. 40%
Hong 2011 [70]RetrospectiveiCCA and pCCA
patients
LT ± neoadjuvant/adjuvant therapy5-yr RFS:
47% (neoadjuvant + adjuvant) vs. 33%
(adjuvant only) vs. 20% (none)
Lunsford 2018 [71]Prospective
case series
Unresectable
iCCA, 6 patients
LT after >6 months disease stability 1-, 3-, 5-yr OS:
100%, 83.3%, 83.3%
RFS: 50%
McMillan 2022 [72]Prospective
case series
follow-up
Unresectable
iCCA, 18 patients
LT after >6 months disease stability1-, 3-, 5-yr OS:
100%, 71%, 57%
1-, 3-yr RFS:
72%, 52%
Semaan 2025 [73]Retrospective
single-center
Unresectable iCCA,
26 patients
Neoadjuvant treatment + LT1-, 3-yr OS: 96%, 82.7%
1-, 3-yr RFS: 70.8%, 56.3%
Yaqub 2025 [74]Prospective
single-center
Unresectable locally advanced iCCA with prior response to
neoadjuvant therapy
LT5 patients underwent LT
2 had recurrence at 12 and 13 months
Teixeira 2024 [79]Case reportUnresectable locally advanced iCCAY90 TARE + Gem/Cis + FOLFOX + LTRecurrence-free at
16-month follow-up
Fernandes 2024 [75]Case reportsUnresectable locally advanced iCCAGemcitabine/cisplatin OR gemcitabine/cisplatin/durvalumab + LTRecurrence-free at
23-month and 6-month
follow-up
Byrne 2025 [76]Case reportUnresectable iCCAY90 TARE + Gem/Cis +
Pemigatinib + LT
Recurrence-free at
1-year follow-up
Ito 2022 [77]Retrospective30 iCCA patientsLT ± neoadjuvant/systemic + LRT1-, 3-, and 5-yr OS:
73%, 46%, 42%
1-, 3-, 5-yr OS (for patients transplanted 2008–2019):
100%, 86%, 69%100% 5-yr OS and RFS (for patients treated with systemic + LRT)
Maspero 2024 [78]Prospective
single-center
13 iCCA patients,
4 patients transplanted
Gem/Cis +
TARE +
LT
5-yr OS: 100% (LT) vs. 0% (no LT)
Table 4. Clinical trials in liver transplant for iCCA.
Table 4. Clinical trials in liver transplant for iCCA.
ReferenceLocationDescriptionNeoadjuvantStudy TypeStatus
NCT02878473 [80]Toronto, Canada5-year overall survival; LT for pts with cirrhosis and unresectable iCCA ≤2 cm confirmed by biopsyNone or
LRT
Multicenter clinical trial,
not
randomized, phase 2
Terminated
NCT04195503 [81]Toronto, Canada5-year overall survival; LDLT for locally advanced unresectable iCCA with no distant mets, LN, or vascular invasion>6 months stability
CTX Alone
Prospective single-centerRecruiting
NCT04556214 [82]Oslo, Norway5-year overall survival; LT for locally advanced unresectable iCCA with no distant mets, LN, or vascular invasion>6 months stability
CTX or LRT
Prospective single-centerRecruiting
NCT06140134 [83]New Jersey, USA5-year overall survival; LT for locally advanced unresectable iCCA with no distant mets, LN, or vascular invasion>6 months stability
CTX ± IO ± TARE
Multicenter clinical trial, not randomized, phase 2Recruiting
NCT06098547 [84]Padova, Italy3-year overall survival; with matched retrospective comparison to CTX alone; LT for locally advanced unresectable iCCA with no distant mets, LN, or vascular invasion>6 months stability
CTX Alone
Prospective single-centerRecruiting
NCT06862934 [85]Milan, Italy3-year overall survival; LT for locally advanced unresectable iCCA with no distant mets, LN, or vascular invasion>6 months stability
CTX+IO+TARE
Prospective single-centerRecruiting
NCT06539377 [86]Jena, Germany5-year overall survival; LDLT for locally advanced unresectable G1/G2 iCCA or HCC/CCA unresectable iCCA with no distant mets, LN, or vascular invasion>6 months stability CTX + TAREProspective single-centerNot yet
recruiting
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lopiano, S.; Guarrera, J.V.; Lunsford, K.E. Therapeutic Advances in Initially Unresectable Locally Advanced Intrahepatic Cholangiocarcinoma: Emerging Treatments and the Role of Liver Transplantation. Curr. Oncol. 2025, 32, 293. https://doi.org/10.3390/curroncol32060293

AMA Style

Lopiano S, Guarrera JV, Lunsford KE. Therapeutic Advances in Initially Unresectable Locally Advanced Intrahepatic Cholangiocarcinoma: Emerging Treatments and the Role of Liver Transplantation. Current Oncology. 2025; 32(6):293. https://doi.org/10.3390/curroncol32060293

Chicago/Turabian Style

Lopiano, Sofia, James V. Guarrera, and Keri E. Lunsford. 2025. "Therapeutic Advances in Initially Unresectable Locally Advanced Intrahepatic Cholangiocarcinoma: Emerging Treatments and the Role of Liver Transplantation" Current Oncology 32, no. 6: 293. https://doi.org/10.3390/curroncol32060293

APA Style

Lopiano, S., Guarrera, J. V., & Lunsford, K. E. (2025). Therapeutic Advances in Initially Unresectable Locally Advanced Intrahepatic Cholangiocarcinoma: Emerging Treatments and the Role of Liver Transplantation. Current Oncology, 32(6), 293. https://doi.org/10.3390/curroncol32060293

Article Metrics

Back to TopTop