Digestive Disease Interventions
DOI: 10.1055/s-0044-1787171
Review Article

Liver Transplantation for Cholangiocarcinoma

Nadine Soliman
1   Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, England
,
Ashton A. Connor
2   Department of Surgery, Houston Methodist Hospital, Houston, Texas
,
Sudha Kodali
3   Department of Medicine, Houston Methodist Hospital, Houston, Texas
,
Rafik Mark Ghobrial
2   Department of Surgery, Houston Methodist Hospital, Houston, Texas
4   Sherrie & Alan Conover Center for Liver Disease and Transplantation, Houston Methodist Hospital, Houston, Texas
5   Center of Immunobiology and Transplantation Sciences, Houston, Texas
6   Department of Surgery, Weill Cornell Medical College, New York, New York
› Author Affiliations
 

Abstract

Cholangiocarcinomas (CCAs) are highly aggressive, primary liver cancers with rising incidence and mortality rates. The current 5-year overall survival is less than 20%. There are no standardized screening protocols, and current diagnostic methods include serum biomarkers and imaging techniques with suboptimal sensitivities and specificities. The most commonly used treatment options, including combination systemic therapies, locoregional therapies, and surgical resection, offer improving but nonetheless limited progression-free and overall survival. Liver transplantation has shown promising results as a potentially curative treatment for two types of CCA, namely, perihilar and intrahepatic. However, the evidence is largely from retrospective series of small to moderate sample sizes. There is a need to define optimal types and sequencing of neoadjuvant and adjuvant peritransplant therapies, as well as criteria for CCA patient transplant eligibility. Here, we conduct a granular review of the evidence available on every step of the transplant care pathway for perihilar and intrahepatic CCA patients. We aim to inform best practices to inform future avenues of research and maximize the number of patients eligible for this potentially life-prolonging therapy.


#

Cholangiocarcinoma (CCA) is an adenocarcinoma of the biliary system. There are three recognized subtypes: perihilar (pCCA, 50–60% of cases), intrahepatic (iCCA, 10–20%), and distal (dCCA, 20–30%).[1] [2] [3] Common risk factors include smoking, chronic inflammation from gallstones, infections (flukes, viral hepatitis), autoimmune conditions, and congenital abnormalities (e.g., choledochal cysts).[4] [5] [6] As these tumors are relatively uncommon and lack effective biomarkers, there is no evidence-based approach to screening. Different management approaches, including surgical, locoregional, and systemic, are used for each subtype with little evidence to inform their most appropriate sequence, resulting in variations in overall survival (OS) and recurrence-free survival (RFS) reported in the literature. Due to the aggressive natural history of CCA, around 65% of patients present at advanced stages or with severe liver dysfunction due to underlying cirrhosis and are therefore ineligible for local therapy options such as percutaneous and endoscopic ablation, local tumor resection (by wedge, minor or major hepatectomy), or radiotherapy.[7] [8] [9] [10] [11] [12] [13] [14] [15] [16] [17] [18] Mortality rates are alarmingly high with reported 5-year OS of less than 20% without intervention.[2] [8] [19] [20] [21] [22] Guidelines recommend margin-negative (R0) liver resection (LR) as the only potentially curative treatment option for iCCA and pCCA with reported OS of 45% and 20 to 40%, respectively. However, in 50 to 70% of iCCA patients, tumor recurrence is seen at a median time of 26 months post-LR.[23] This has led hepatobiliary and transplant specialties to explore other treatment modalities, specifically liver transplantation (LT), with more recent studies showing promising results. Historically, LT for CCA had poor RFS and OS, with early studies reporting a 5-year OS of only 20% in iCCA patients.[2] [8] [24] [25] [26] [27] [28] [29] Yet, recent evidence has shown improved outcomes in those with small, early-stage tumors.[30] [31] When combined with neoadjuvant chemotherapy, post-LT 5-year OS for patients with iCCA improved up to 78%, and 5-year RFS up to 65% in pCCA patients treated with neoadjuvant chemoradiotherapy.[14] [32] [33] [34] [35] This suggests that careful selection and stratification of CCA patients based on prognostic clinicopathological factors such as tumor size, burden, multifocality, response to neoadjuvant therapy, and lymphovascular invasion would increase the number of iCCA and pCCA patients eligible for LT as a potentially curative therapy and will be the focus of this review.[3] [31] [36] [37] [38] [39] This is also summarized in [Fig. 1].

Zoom Image
Fig. 1 Integration of standard liver transplant care (top row) with cholangiocarcinoma-specific medical approaches (bottom row) to optimize liver transplantation process.

Diagnosis, Referral, and Evaluation

The pretransplant evaluation process for patients with CCA is variable, beginning with a referral to a transplant center often due to underlying primary sclerosing cholangitis (PSC) or liver dysfunction (cirrhosis), and sometimes prior to a confirmed diagnosis of malignancy. This is in part as patients with CCA are typically asymptomatic until advanced stages. After referral to a transplant center, patients are evaluated by a tumor board composed of a multidisciplinary team (including hepatology, medical oncology, surgery, and radiation oncology among others), where recommendations given by cardiopulmonary specialists and psychosocial assessments are combined with a thorough review of imaging studies to assess patient eligibility for transplant and to recommend personalized neoadjuvant regimens. Not all transplant centers have developed this integrated, subspecialized care, and a possible volume–outcome relationship has been suggested in the literature.[40] There are no standardized screening protocols for CCA due to possible need for invasive procedures and a lack of promising evidence found in prospective series.[41] Diagnosis of CCA using noninvasive methods is challenging owing to difficulties accurately distinguishing CCA from other primary liver cancers. Currently, the only accepted serum biomarker for diagnosis and screening is CA19–9, with known sensitivity and specificity of 60 to 93% and 78 to 98%, respectively, in patients with underlying PSC and around 75 and 80%, respectively, in the absence of PSC.[42] [43] [44] [45] [46] [47] [48] [49] [50] [51] However, studies have previously reported sensitivities as low as 33% in patients with resectable CCA, suggesting limited value to the use of serum CA 19–9 levels in identifying patients with early stage, surgically resectable disease.[50] Imaging with modalities such as ultrasound, computed tomography (CT), and magnetic resonance imaging (MRI) with or without contrast have also been suggested as tools in the screening and diagnosis of CCA.

In patients with underlying cirrhosis undergoing screening for HCC, iCCA may be detected incidentally at early stages. One large retrospective study identified incidental iCCA in 1% of explants from LT patients suspected of having HCC (23 out of 2,301),[52] though the exact rate of incidental iCCA found on liver explant remains unclear. There are difficulties in radiologically distinguishing between the two primary malignancies, leading to cases of confirmed diagnosis only after LR and/or transplantation on final explant pathology, a concerning occurrence due to rising promising evidence in favor of neoadjuvant systemic therapy for iCCA.[3] [53] [54] [55] In around 30 to 42% of cases, PSC-associated CCA was reported to be found incidentally on autopsy or on liver explants post-OLT.[56] [57] [58] While MRI can provide a more comprehensive assessment of the primary mass, CT is more suited for the detection of hepatic vasculature and is therefore critical in determining resectability.[59] 18F-fluorodeoxyglucose (18F-FDG) PET imaging has shown poor performance in the detection of primary tumors but surprisingly high sensitivity and specificity for the detection of lymph node and distant metastasis and, therefore, is important in the staging of all CCA subtypes.[60] [61]

In patients with pCCA and dCCA, initial CT features may warrant additional imaging with magnetic resonance cholangiopancreatography (MRCP), which has a sensitivity and specificity of 87 and 85%, respectively, in differentiating between benign and malignant causes of hilar obstruction.[62] [63] MRCP also has an additional benefit of being able to map a patient's biliary anatomy prior to endoscopic intervention with ERCP. ERCP is employed as a diagnostic and therapeutic tool as it enables the detection of malignant strictures and acquisition of biliary brushings for cytology and confirmation of the underlying tumor biology.[19] However, one meta-analysis examining the use of biliary brushings for pCCA detection found a low sensitivity of only 43%.[64] Endoscopic ultrasonography (EUS) has been previously recommended in the diagnosis and staging of pCCA owing to its ability to provide a detailed assessment of the extrahepatic bile duct and concurrent tissue acquisition via fine-needle aspiration (FNA).[19] However, EUS-FNA has demonstrated a higher sensitivity for dCCA than for pCCA (81 vs. 59%), with controversies arising concerning the potential risk of tumor dissemination associated with EUS-guided tissue biopsy of pCCA.[19] [65] [66]

While there are many tools available for the diagnosis and staging of CCA ([Fig. 1]), overall, these have shown subpar sensitivity and specificity, suggesting the need for further investigations aimed at finding standardized, effective methods involving a combination of these protocols, primarily aimed at confirming CCA diagnosis pretransplant to best guide neoadjuvant therapy.


#

Prognostic Factors and Waitlist Selection

Although LT is becoming one of the most promising treatments for CCA, concerns regarding national organ shortages combined with early studies demonstrating poor prognosis for iCCA patients' post-LT has led to hesitancy in incorporating these cancers as formal indications in the United Network for Organ Sharing (UNOS) standards. While this was previously true for all CCA subtypes, a study conducted by the Mayo Clinic showed favorable results in patients with pCCA comparable to benign indications for LT. UNOS subsequently granted an exception to the Model for End Stage Liver Disease (MELD), allowing patients who had suitable tumor size and met other diagnostic criteria to be added to the waiting list for LT with MELD scores equivalent to those of a patient with stage 1 HCC (an existing indication for LT). This resulted in a greater number of patients having access to LT.[67] [68] The standard MELD exception point for pCCA is set at Median MELD at transplant (MMAT) −3 points.[69] A patient must have unresectable disease either due to locally advanced tumor with vascular and/or biliary invasion preventing R0 resection, or poor functional reserve due to underlying liver disease to qualify for MELD exception points.[69] [70] There must also be only a single tumor <3 cm in diameter with no intra- or extrahepatic metastasis seen, and patients must be treated with neoadjuvant therapy at centers with approved protocols.[70] Currently, there are no formal UNOS indications for LT for curative intent in patients with iCCA, and hence no established, standardized inclusion criteria. A recent propensity score–matched National Cancer Database (NCDB) study has shown that neoadjuvant chemotherapy was associated with longer OS in select patients with CCA compared with those directly undergoing surgical resection with subsequent adjuvant chemotherapy[71] confirming results seen in studies of CCA patients pretransplant,[72] [73] [74] and suggesting the need for further studies looking at tailored neoadjuvant therapy to improve the tumor response rate and increase the number of patients eligible for LT.

Historically, for patients with pCCA, registry-based LT outcomes were poor,[26] even for small tumors identified incidentally on explant.[75] Improved outcomes of 5-year RFS of 65% were seen with the introduction of neoadjuvant chemo-radiotherapy for systemic control prior to LT.[32] [33] [34] [35] This suggested that the use of biology-based selection criteria for LT may result in more favorable outcomes than those seen with the use of size or burden-based criteria alone.[3] Many transplant centers within the United States, and internationally, now use the Mayo Clinic protocol of chemoradiation prior to LT for the treatment of unresectable pCCA with few deviations.[76] This protocol involves pretreatment with radiation and 5FU followed by brachytherapy with iridium and concomitant 5FU followed by maintenance single-agent chemotherapy until LT. A recent meta-analysis of retrospective series of LT for pCCA reported improved 5-year OS of 51.7% and 3-year RFS of 51.7% in those given neoadjuvant therapy compared with those without (5-year OS: 31.6%, 3-year RFS: 24.1%), confirming the importance of neoadjuvant therapy in the pre-LT period. However, patient dropout rate during neoadjuvant therapy cited in the literature ranges from 10 to 66.7%, with common causes including disease progression (41%) and death prior to transplantation (10.7%).[76] [77] Following advances in systemic therapy for other cancers, including pancreatic and rectal, the continued role of radiotherapy in those neoadjuvant settings prior to surgery has been called into question, and may require further investigation to determine true benefit.[78] [79]

Historically, for patients with iCCA, LT without adjunctive treatment has dismal outcomes, with early studies showing 1- and 3-year OS of only 19.4 to 38% and 4.9 to 10%, respectively. This resulted in iCCA becoming a relative contraindication for LT.[80] [81] More recent studies have challenged this, showing improved outcomes (1- and 3-year OS rates of 83.3–100% and 47.91–83.3%, respectively) with improved patient selection and standardized use of neoadjuvant therapy ([Table 1]).[22] [31] [36] [37] [38] [39] [82] [83] [84] [85] [86] [87] [88] In a study conducted by Hong et al, 38 CCA cases (iCCA and pCCA) received LT with significant differences seen in 5-year OS (47, 33, and 20%, p = 0.03) in patients who received neoadjuvant and adjuvant, adjuvant therapy only, or no therapy, respectively.[82] The neoadjuvant protocol consisted of transarterial chemoembolization or radiotherapy combined with chemotherapy. In 2014, a multicenter retrospective cohort reported 1-, 3-, and 5-year OS rates of 93, 83, and 65%, respectively, and low recurrence rates for 48 LT patients with existing cirrhosis and small (<2 cm) incidental iCCA.[31] These outcomes are similar to those achieved in patients transplanted for well-selected hepatocellular carcinoma and are superior to those seen with local resection (LR). In 2022, University of California, Los Angeles (UCLA) reported 1-, 3-, and 5-year OS rates of 80, 63, and 49%, respectively, for CCA patients undergoing LT.[39] [89] Studies have reported strong associations between poor tumor differentiation, microvascular invasion, and worse post-LT outcomes.[90] [91] [92] Researchers at UCLA established a prognostic scoring system that has been shown to correlate with LT outcomes in iCCA patients.[36] [84] Predictive risk factors included lack of neoadjuvant or adjuvant therapy, multifocality, infiltrative growth, history of PSC, and perineural and lymphovascular invasion.[36] Considering these factors, the team at Houston Methodist reported prospective case series of patients with unresectable iCCA treated with LT and neoadjuvant chemotherapy with promising results of 1-, 3-, and 5-year OS rates of 100, 83, and 83%, respectively, in the first study[37] and 100, 71, and 57%, respectively, in the second study.[38] Patients in these studies who were listed but not transplanted had a decline in survival after 1 year, with no patients alive within 2 years, outcomes consistent with those seen in previous iCCA patients treated with only systemic therapy.[93] The relative scarcity of iCCA LT series that employ neoadjuvant therapy impedes development of standardized treatment algorithms. Recent series from Houston Methodist has shown excellent LT outcomes in iCCA patients treated with neoadjuvant gemcitabine and cisplatin without radiation with 1-year OS of 100% and 5-year OS of 75%.[94] These results correspond to those at UCLA[39] and demonstrate that LT is a potentially curative treatment option in well-selected iCCA patients.

Table 1

OS and RFS rates for CCA subtypes with and without LT

CCA subtype

OS

RFS

Without LT

With LT

Without LT

With LT

pCCA

20–40%[172]

82.8%[76]

50–70%[12] [173] [174]

75.9%[76]

iCCA

45%

75%[109]

30–50%[23]

70%[109]

dCCA

48–52%[175] [176]

NA

∼48%[175] [176]

NA

Abbreviations: dCCA, distal cholangiocarcinoma; CCA, cholangiocarcinoma; iCCA, intrahepatic cholangiocarcinoma; LT, liver transplantation; OS, overall survival; pCCA, perihilar cholangiocarcinoma; RFS, recurrence-free survival.


The utilization of other neoadjuvant therapies such as stereotactic body radiation therapy (SBRT) has shown promise in preventing disease progression with lower toxicity than traditional radiotherapy.[95] [96] Recent studies have found that neoadjuvant selective internal radiotherapy with Y90 either alone or combined with systemic chemotherapy can be used to bridge or downstage patients with unresectable iCCA tumors to resection with good survival outcomes, disease control, and an acceptable safety profile, although not specifically in the transplant setting.[97] [98] [99] [100] [101] [102] [103] Advances in oncological therapy have led to improved management of CCA with greater survival seen with the combination of folinic acid, fluorouracil, and oxaliplatin (FOLFOX) as a second-line chemotherapy agent for use as either neoadjuvant or adjuvant therapy in CCA patients considered for LT.[95] [104] Promising results have also been seen in trials testing immunotherapy for CCA; however, more research is needed before these treatments can be routinely recommended in the peritransplant setting.[95] [105]

Two possible approaches to patient selection currently used include burden-based and biology-based. Initial studies advocated for selection based on iCCA size and number,[31] and later studies demonstrated promising outcomes in patients selected based on response to neoadjuvant therapy, independent of tumor size.[37] [38] [39] [87] Following the success of patient selection based on tumor burden for HCC and pCCA, most centers perform LT only for patients with small iCCA tumor burden, supported by reports from Mount Sinai Medical Center and Sun Yat-sen University.[106] [107] [108] A recent meta-analysis found similar results and agree that smaller tumor size is associated with improved OS.[106] [109] However, using size-based criteria limits LT to a small percentage of iCCA patients with very small tumors, with most patients being ineligible to receive potentially lifesaving treatment. An international retrospective study showed that tumor size did not independently predict tumor recurrence.[31] Later studies supported these results and showed that tumor burden is not an accurate predictor of patient outcomes after LT.[39] [110] Shifting toward biology-based selection criteria has shown promise. With reports from Houston Methodist Hospital showing that tumor response to neoadjuvant therapy is a better predictor of OS than tumor size.[37] [38] [106] Additionally, recent advances in next-generation sequencing (NGS) of solid tumor biopsies and liquid biopsy of cell-free DNA, RNA, and/or protein expression is promising in the pre-LT identification of tumor genetic mutations responsive to neoadjuvant therapies and capable of predicting patient prognosis to optimize selection of patients amenable to LT. Identification of these mutations could also assist in personalizing therapies, surveillance, and to predict and treat recurrence.[3] [7] [111] Studies of patients receiving LT for iCCA have identified mutations in genes such as FGFR2 and IDH1 which have approved targeted therapies.[38] [112] Worse overall prognosis in iCCA patients has been observed with intratumoral heterogeneity; however, more research is needed to ascertain its significance in predicting post-LT outcomes.[113] [114] [115] [116] An important factor to consider when determining selection criteria is that excessive expansion of inclusion criteria will likely result in a significant increase in organ demand, at the time of organ shortages, with potential decreases in OS due to increased waiting times among all patients on the waitlist irrespective of underlying hepatic malignancy.[117]


#

Liver Transplantation

With a current shortage in organs, optimization of the LT procedure for CCA remains of the utmost importance to minimize risk of rejection, graft failure, or tumor recurrence.

A recent meta-analysis of patients with iCCA found pooled 1-, 3-, and 5-year OS rates of 75, 56, and 42%, respectively.[109] The pooled 1-, 3-, and 5-year RFS rates were 70, 49, and 38%, respectively, with patients with underlying cirrhosis showing higher RFS rates.[109] Superior 5-year RFS rates were seen in patients with very early (single ≤2 cm) iCCA (67%) compared with patients with advanced iCCA (34%).[109] Another meta-analysis comprising 20 studies and 428 patients looked at pooled OS and RFS rates following LT for pCCA patients.[76] They found 1-, 3-, and 5-year OS in LT patients without neoadjuvant therapy to be 71.2, 48.0, and 31.6%, respectively. These improved to 82.8, 65.5, and 65.1% in patients with completed neoadjuvant chemoradiotherapy regimens.[76] Three-year recurrence rates were reported to be 24.1% in patients given neoadjuvant chemoradiotherapy, and 51.7% in those without. Interestingly, patients with PSC seemed to have the most favorable outcomes.[76]

Currently, there remains a poor accuracy of preoperative imaging assessment and models to identify patients at high risk of nodal disease. In patients with small iCCA, nodal status was found to be the main determinant of prognosis with 5-year OS reported to be 0 to 20% in N1 patients compared with 35 to 50% in N0 patients.[118] [119] [120] [121] Predicting LN size by CT or MRI has shown poor PPV of only 2.8 to 48%, possibly due to nodal enlargement secondary to reactive hyperplasia.[120] This led to a focus on 18FDG-PET for nodal staging which showed an accuracy of 81% for size >1 cm LNs, and a sensitivity and specificity of 69.1 and 88.4%, respectively, making it highly predictive for nodal involvement.[61] [120] [122] EUS with fine needle aspiration (FNA) is increasingly being used in clinical practice preoperatively to assess the presence of nodal disease; however, few studies are available in the literature to recommend routine use.[120]

Some studies have recently investigated the effect of pretransplant lymphadenectomy on OS rates in iCCA patients. Yet, it remains a topic of ongoing debate. The 8th edition of the TNM staging system proposed by the AJCC highlighted that hepatic pedicle lymphadenectomy according to tumor location is recommended to ensure precise staging.[123] Regional LNs associated with iCCA are defined as inferior phrenic, hilar, and gastrohepatic LNs for left liver iCCAs (draining stations 12 to 8, and 7, 1, 3) and hilar, periduodenal, and peripancreatic LNs for right liver iCCAs (draining stations 12 to 8, and 13). Spread of the tumor to the celiac, periaortic, and/or pericaval LNs is considered M1 disease irrespective of primary location.[22] [120] [123] Despite the AJCC's recommendation of a minimum of six harvested lymph nodes (HLNs) for adequate nodal staging, routine lymphadenectomy with histologic examination is poorly practiced.[124] [125] [126] A study by Bagante et al found that 5-year OS in N0 patients improved with an increased number of HLNs, 54.9% in patients with six or more HLNs versus 39.4% in patients with less than six HLNs. Of note, patients with less than six nodes harvested also tended to have an increased risk of death compared with those with six or more HLNs (hazard ratio: 1.39).[126] No significant change was observed in patients with N1 disease, irrespective of number of lymph nodes harvested (p = 0.71); however, only 25% of patients in the study had adequate nodal staging performed.[126] The 5-year OS of patients with negative lymph nodes (NLNs) was 44.4 versus 15.2% for patients with metastatic lymph nodes (MLNs) (p < 0.001), illustrating the importance of adequate nodal staging in predicting patient prognosis and optimizing pretransplant patient risk factors. In patients with positive N1 iCCA, Kim et al found that expanding dissection to station nos. 12 and 8 covered 82.0% (n = 50) of metastatic cases.[127] In patients with cN0 disease, a study by Sposito et al found that adequate lymphadenectomy provided better survival outcomes for patients with cN0 disease found to be N positive on pathology with longer OS (28 vs. 23 months) and DFS (13 vs. 9 months).[128] This supports the push for more surgeons to use adequate lymphadenectomy as routine practice even in patients with cN0 iCCA.

In pCCA patients, lymph node metastasis is common, seen in 31 to 58% of patients, likely due to a thin bile duct wall and remains one of the most important prognostic factors.[129] [130] [131] The most common sites of metastasis are around the bile duct (27.1–42.7%), portal vein (30.9–35.7%), common hepatic artery (27.3–31.3%), para-aorta (17.3%), posterior pancreatic head (14.5–50%), and celiac trunk (6.4–14.3%).[132] [133] Mantel et al reported that in N0 patients, the 5-year OS of patients with MLNs was significantly lower than those without (27 vs. 54%, p = 0.01), and not significantly different from N1 patients (27 vs. 15%, p = 0.54).[129] [134] As a result, the Japan Society of Hepatobiliary and Pancreatic Surgery recommends dissection of the first and second stations (stations 8, 12, and 13) for pCCA differing from the National Comprehensive Cancer Network (NCCN) guidelines that suggest standard lymph node dissection of stations 12 and 13 with any further metastasis considered a contraindication to radical surgery.[135] [136] A study by Kitagawa et al found that 3- and 5-year OS rates were 31.8 and 14.6% for patients without lymph node metastasis, 31.8 and 14.7% for patients with regional lymph node metastases, and 12.3 and 12.3% for patients with para-aortic lymph nodes metastases.[132] Of the patients with para-aortic lymph node metastases, seven patients had no obvious signs of lymph node involvement during surgery and was confirmed by postoperative pathology examination. The outcomes of these patients were significantly better than those with lymph node metastases confirmed intraoperatively and were equal to that of patients with regional lymph node metastases.[129] [132] This suggests that even in patients with para-aortic MLNs, extended lymphadenectomy can provide better outcomes with no increased risk of procedural complications.[13] [132] [137] [138] The 5-year OS rates of pCCA with regional lymphadenectomy were 7 to 20% versus 26 to 46% in pCCA patients with extended lymphadenectomy, supporting the value of extended lymphadenectomy in resectable pCCA.[139] [140] [141] [142] [143] [144] [145] Ma et al found that extended lymphadenectomy significantly increases lymph node retrieval, reducing risk of understaging, improving prognosis predictions, and OS in patients with M0 diseases with R0 resection.[146] However, no OS benefit was found in patients with M1 disease, concluding that extended lymphadenectomy should not be performed in pCCA patients with intraoperatively confirmed distant MLNs.[129] Some studies found no significant difference in OS between patients with N1 and N2 MLNs, challenging the accuracy of the AJCC staging system which relies on site of lymph node metastasis.[131] [139] [147]

Intraoperatively, there remains some debate regarding whether Roux-en-Y choledochojejunostomy or duct-to-duct choledochocholedochostomy for biliary reconstruction provided better outcomes. No studies were done directly comparing these methods in the context of LT for CCA. One meta-analysis looking at LT in PSC patients found no significant differences in anastomotic bile leak rates, graft survival, PSC recurrence, and incidence of de novo CCA following transplantation.[148] Another study comparing both methods in all patients receiving right lobe living donor LT (LDLT) irrespective of indication found an increased incidence of stricture but a significantly lower incidence of leakage with duct-to-duct choledochocholedochostomy, with 74.5% of the strictures later managed with endoscopic treatment.[149] Overall consensus recommends duct-to-duct choledochocholedochostomy when feasible.

There is also debate on the use of LDLT versus deceased donor liver transplant (DDLT) on patient outcomes. Studies have shown that longer time elapsed between neoadjuvant therapy and LT leads to decreased incidence of recurrence. However, patients with prolonged intervals may develop radiation-induced fibrosis which could complicate the staging and operative process.[73] This may be in part solved with LDLT by removing waitlist for a deceased donor and aiding physicians in optimizing operative timings. Series have shown no significant differences in outcomes post-LT for PSC-associated pCCA in patients receiving LDLT versus DDLT.[150] LDLT for de novo pCCA showed trends toward increased recurrence and worse OS when compared with DDLT, though the differences were not statistically significant. Researchers are more focused toward understanding the underlying mechanism of disease progression after neoadjuvant treatment to better select patients and prevent posttransplant recurrences.[151] Another study confirmed these findings by suggesting that while LDLT may offer shorter waiting times, it is associated with higher risks of biliary complications compared with DDLT (34 vs. 17%, p < 0.001).[152] Using extended criteria donors of advanced age, steatosis or circulatory death may increase the number of organs available but is associated with higher 3-year graft failure rates of 27.3% for DCD livers versus 18.2% for donation after brainstem death.[95] [153] A possible solution involves the use of normothermic machine perfusion to evaluate graft function preoperatively and help select grafts with optimal function thereby increasing the donor pool and improving patient outcomes.[154]

These considerations have led to consensus criteria for exception points for LT in the United States for pCCA. Transplant centers should have an approved written protocol outlining selection criteria, neoadjuvant therapy, and operative staging protocols. Patients should be deemed unresectable at multidisciplinary cancer conference, have cross-sectional imaging demonstrating a single lesion less than 3 cm in maximum size without extrahepatic spread, and have no lymph node or peritoneal involvement on operative staging after completion of neoadjuvant therapy.[155] There are no adopted LT consensus criteria for iCCA, but American Association for the Study of Liver Disease (AASLD) guidelines suggest that unresectable solitary tumors up to 5 cm with stability or response to neoadjuvant therapy can be considered for LT under institutional research protocols.[156]

Post-LT outcomes may also vary with transplant center expertise. A database study of all LT patients with CCA in the United States found low center volume to be associated with worse post-LT OS and graft survival, while a multicenter study of LT for pCCA found equivalent outcomes irrespective of center volume.[3] [35] [40] [109]


#

Post-LT Follow-up

Currently, there are no standardized approaches to follow-up protocols for patients with CCA post-LT. The length of follow-up, need for scans, blood tests, or adjuvant chemotherapy is center specific with no clear evidence or high-quality studies performed. The recent BILCAP study provided the best evidence for the use of adjuvant capecitabine post-LR with median OS reported as 53 months in the capecitabine group versus 36 months in the observation group, and median RFS was 25.9 months in the capecitabine group and 17.4 months in the observation group with prespecified per-protocol analysis.[157] However, no evidence was provided for patients >70 years of age, and the trial was not able to meet its primary endpoint of improved OS in the intention-to-treat population. A follow-up publication of the same trial focusing on intention-to-treat analysis found median OS of 49.6 months in the capecitabine group compared with 36.1 months in the observation group, further supporting the previous study and suggesting that capecitabine can be used as adjuvant chemotherapy after surgery to improve OS in patients with resected CCA and should be considered the standard of care.[158] One meta-analysis showed statistically significant improvements in OS in patients receiving chemotherapy or chemoradiotherapy compared with those receiving radiotherapy alone (OR: 0.39, 0.61, and 0.98, respectively; p = 0.02), with the greatest benefit seen in patients with LN+ and R1 disease.[159] THE SWOG S0809 study found that chemotherapy and radiotherapy post-LR may increase OS and decrease rates of local recurrence citing a 2-year OS of 65%.[160] Jeong et al conducted a randomized controlled study comparing outcomes in patients with pCCA or dCCA divided into two groups, one receiving adjuvant gemcitabine and cisplatin (GemCis) and the other receiving adjuvant capecitabine, with no statistically significant differences in OS and DFS reported.[161] No studies were conducted looking at the effect of adjuvant capecitabine after LT. Therefore, more research is required to ascertain whether improved survival would be found in patients undergoing LT for CCA.

There is a shortage of studies in the literature focusing on optimal length of follow-up for patients with CCA. Some authors have based their clinical decisions on presenting symptoms, physical examinations, serum CEA and CA19–9, and CT scan results.[162] [163] [164] Rizzo et al published a 20-year retrospective study focused on this topic where biliary tract cancers (BTC) patients were followed up every 3 months during the first 2 years post-LR and every 6 months from the third to the fifth postoperative year. At each follow-up visit, the patients were examined, blood work was obtained (CEA and CA 19–9), and an abdominal/chest CT scan with IV contrast was performed.[164] Results suggest that intensive follow-up after surgical resection should be implemented to help identify disease relapse and allow for early treatment and prolonged survival in such cases.[164]

Few studies have been conducted investigating the use of NGS, molecular profiling, and minimal residual disease (MRD) on cancer surveillance and recurrence in CCA patients. A study by Lamarca et al showed a trend toward increased risk of recurrence in patients with pancreatic and biliary tract malignancies when circulating tumor DNA (ctDNA) was present after LR; however, the results were not statistically significant.[165] Another study attempted to stratify iCCA patients post-LR for risk of recurrence and OS using clinical variables and tumor sequencing.[166] Patients were stratified into low-risk (solitary nodules, LN − ) and high-risk (multifocal and/or LN + ) categories and further divided by the presence or absence of mutations in TP53, KRAS, and/or CDKN2A.[166] The presence of these mutations was independently associated with worse patient prognosis. More research is needed to determine the clinical utility of NGS and ctDNA analysis in the post-LT follow-up period to assess risk of recurrence and the presence of MRD.


#

Future Directions

Advances made in the past few years have improved patient outcomes after LT for CCA. Additional studies are needed to validate these findings and aid in the development of standardized protocols. Biomarkers have shown promise in potentially prognosticating patient survival post-LT and predicting response to neoadjuvant therapy, leading to better outcomes overall. Additional research into these biomarkers would allow for noninvasive prediction of post-LT outcomes, identification of aggressive tumor subtypes, and prioritization of LT in patients with a low risk of recurrence.[106] Additionally, these biomarkers could be used in the peri-LT period to tailor neoadjuvant and adjuvant targeted therapies to the individual patients' tumor genetic profile, further optimizing patient posttransplant survival and improving LT candidate selection criteria by increasing response to neoadjuvant therapy. Further basic and translational research in drug development is needed to create novel treatments for CCA tumors that are not responsive to current therapies.[106] [167]

At present, LT in CCA literature consists mainly of retrospective case series, with heterogeneous patient populations with respect to disease stage, neoadjuvant therapies, LT donor type, and postoperative management. Ideally, more prospective multicenter observational studies or randomized controlled trials are needed for evaluation of the effectiveness of the currently employed protocols and whether any modifications could be favorable. However, establishing these trials remains challenging owing to the rarity of CCA, aggressiveness of the tumor subtypes, and difficulties in accurately diagnosing and screening patients, among other confounding factors that act as obstacles in the recruitment of suitable patients using standardized inclusion criteria to achieve a homogenous patient population. This could be aided, in part, through the development of international registries to increase the number of cases and providing a platform for the systematic collection of relevant variables. An ongoing trial since 2014, TRANSPHILL in France, aimed to recruit 54 patients with pCCA for randomization to either curative resection or LT, with primary outcomes of 3-year RFS and 5-year OS. Results are still pending and could represent a shift toward increased use of LT for curative intent in patients with pCCA.[95] [168]

Advancements in neoadjuvant therapies such as SBRT and newer chemotherapy agents have shown promising results in reducing disease progression. SBRT has been shown to have lower toxicity than traditional external beam radiotherapy and therefore may become the preferable treatment pre-LT.[96] A recently reported combination of folinic acid, fluorouracil, and oxaliplatin (FOLFOX) has shown improvement in survival when used as a second-line chemotherapy agent, with recent studies also advocating for the benefits of immunotherapy in CCA.[104] [105] The BILCAP trial suggested capecitabine as an effective adjuvant therapy post-LR for CCA, citing improved patient OS rates; however, studies are needed in the transplant setting to see if similar results are found.[157] [158]

The use of immunosuppression, however, is known to increase the risk of recurrence and development of malignancy. Therefore, there needs to be an optimized regimen established and a balance found between the risk of graft rejection and tumor recurrence after LT for CCA. A recent study conducted on iCCA and pCCA patients undergoing LT found that a reduced dose of immunosuppressives was associated with a significantly increased odds ratio of survival after recurrence (4.2, p = 0.02).[169]

Future research into novel biological agents and chemotherapy regimens will result in better selection of patients for LT, better personalization of neoadjuvant and adjuvant therapies, and improved patient outcomes.[170]

For the standardized establishment of LT as an effective curative treatment option for CCA, iCCA needs an established indication for MELD exception, and both iCCA and pCCA literature require consistent reports of more than 50% 5-year survival rates for patients selected using inclusion criteria consistent with existing established LT indications. There will also be a need for increased supply of donor livers to match the increased demand that may, in part, be solved through improvements in technologies such as normothermic machine perfusion and changes to policies such as shifts toward opt-out organ donation.[69] [106] [171]


#

Conclusion

In conclusion, LT is a promising curative treatment for pCCA and iCCA, showing improved OS in most patients. However, more research is needed to establish standardized selection criteria to ensure fair access of a greater number of patients to LT. Much of the evidence on peri-LT care is borrowed from LR, locoregional, and systemic therapy literature. Additionally, the field is currently lacking consensus on post-LT follow-up protocols, specifically the length of follow-up time, type of imaging or blood tests used, and use of adjuvant therapy, an important step in the optimization of patient treatment. While neoadjuvant chemoradiotherapy has been associated with improved outcomes, more studies should be conducted looking at tailoring these treatments, perhaps with the use of NGS and molecular profiling, to each patient's tumor subtype. Overall, there is a lack of high-quality studies present in the literature most likely due to a heterogeneous patient population and a lack of consistency in variables collected that may in part be solved with the establishment of national and international databases aimed at reducing confounding factors and center bias. Owing to the current organ shortage, strict indications and contraindications to LT are necessary alongside existing efforts to improve the national organ supply.


#
#

Conflict of Interest

None declared.

  • References

  • 1 Loeuillard E, Conboy CB, Gores GJ, Rizvi S. Immunobiology of cholangiocarcinoma. JHEP Rep Innov Hepatol 2019; 1 (04) 297-311
  • 2 Rizvi S, Khan SA, Hallemeier CL, Kelley RK, Gores GJ. Cholangiocarcinoma - evolving concepts and therapeutic strategies. Nat Rev Clin Oncol 2018; 15 (02) 95-111
  • 3 Connor AA, Kodali S, Abdelrahim M, Javle MM, Brombosz EW, Ghobrial RM. Intrahepatic cholangiocarcinoma: the role of liver transplantation, adjunctive treatments, and prognostic biomarkers. Front Oncol 2022; 12: 996710
  • 4 Rizvi S, Gores GJ. Pathogenesis, diagnosis, and management of cholangiocarcinoma. Gastroenterology 2013; 145 (06) 1215-1229
  • 5 Clements O, Eliahoo J, Kim JU, Taylor-Robinson SD, Khan SA. Risk factors for intrahepatic and extrahepatic cholangiocarcinoma: a systematic review and meta-analysis. J Hepatol 2020; 72 (01) 95-103
  • 6 Tyson GL, El-Serag HB. Risk factors for cholangiocarcinoma. Hepatology 2011; 54 (01) 173-184
  • 7 Abdelrahim M, Esmail A, Abudayyeh A. et al. Transplant oncology: an evolving field in cancer care. Cancers (Basel) 2021; 13 (19) 4911
  • 8 Banales JM, Marin JJG, Lamarca A. et al. Cholangiocarcinoma 2020: the next horizon in mechanisms and management. Nat Rev Gastroenterol Hepatol 2020; 17 (09) 557-588
  • 9 Weber SM, Ribero D, O'Reilly EM, Kokudo N, Miyazaki M, Pawlik TM. Intrahepatic cholangiocarcinoma: expert consensus statement. HPB (Oxford) 2015; 17 (08) 669-680
  • 10 DeOliveira ML, Cunningham SC, Cameron JL. et al. Cholangiocarcinoma: thirty-one-year experience with 564 patients at a single institution. Ann Surg 2007; 245 (05) 755-762
  • 11 Jarnagin WR, Fong Y, DeMatteo RP. et al. Staging, resectability, and outcome in 225 patients with hilar cholangiocarcinoma. Ann Surg 2001; 234 (04) 507-517 , discussion 517–519
  • 12 Kobayashi A, Miwa S, Nakata T, Miyagawa S. Disease recurrence patterns after R0 resection of hilar cholangiocarcinoma. Br J Surg 2010; 97 (01) 56-64
  • 13 Kosuge T, Yamamoto J, Shimada K, Yamasaki S, Makuuchi M. Improved surgical results for hilar cholangiocarcinoma with procedures including major hepatic resection. Ann Surg 1999; 230 (05) 663-671
  • 14 Rea DJ, Heimbach JK, Rosen CB. et al. Liver transplantation with neoadjuvant chemoradiation is more effective than resection for hilar cholangiocarcinoma. Ann Surg 2005; 242 (03) 451-458 , discussion 458–461
  • 15 Rea DJ, Munoz-Juarez M, Farnell MB. et al. Major hepatic resection for hilar cholangiocarcinoma: analysis of 46 patients. Arch Surg 2004; 139 (05) 514-523 , discussion 523–525
  • 16 Su CH, Tsay SH, Wu CC. et al. Factors influencing postoperative morbidity, mortality, and survival after resection for hilar cholangiocarcinoma. Ann Surg 1996; 223 (04) 384-394
  • 17 Washburn WK, Lewis WD, Jenkins RL. Aggressive surgical resection for cholangiocarcinoma. Arch Surg 1995; 130 (03) 270-276
  • 18 Spolverato G, Bagante F, Tsilimigras D, Ejaz A, Cloyd J, Pawlik TM. Management and outcomes among patients with mixed hepatocholangiocellular carcinoma: a population-based analysis. J Surg Oncol 2019; 119 (03) 278-287
  • 19 Brindley PJ, Bachini M, Ilyas SI. et al. Cholangiocarcinoma. Nat Rev Dis Primers 2021; 7 (01) 65
  • 20 Tomlinson JL, Valle JW, Ilyas SI. Immunobiology of cholangiocarcinoma. J Hepatol 2023; 79 (03) 867-875
  • 21 Lee H, Ross JS. The potential role of comprehensive genomic profiling to guide targeted therapy for patients with biliary cancer. Therap Adv Gastroenterol 2017; 10 (06) 507-520
  • 22 Mazzaferro V, Gorgen A, Roayaie S, Droz Dit Busset M, Sapisochin G. Liver resection and transplantation for intrahepatic cholangiocarcinoma. J Hepatol 2020; 72 (02) 364-377
  • 23 Hyder O, Marques H, Pulitano C. et al. A nomogram to predict long-term survival after resection for intrahepatic cholangiocarcinoma: an Eastern and Western experience. JAMA Surg 2014; 149 (05) 432-438
  • 24 Meza-Junco J, Montano-Loza AJ, Ma M, Wong W, Sawyer MB, Bain VG. Cholangiocarcinoma: has there been any progress?. Can J Gastroenterol 2010; 24 (01) 52-57
  • 25 Casavilla FA, Marsh JW, Iwatsuki S. et al. Hepatic resection and transplantation for peripheral cholangiocarcinoma. J Am Coll Surg 1997; 185 (05) 429-436
  • 26 Meyer CG, Penn I, James L. Liver transplantation for cholangiocarcinoma: results in 207 patients. Transplantation 2000; 69 (08) 1633-1637
  • 27 Shimoda M, Farmer DG, Colquhoun SD. et al. Liver transplantation for cholangiocellular carcinoma: analysis of a single-center experience and review of the literature. Liver Transpl 2001; 7 (12) 1023-1033
  • 28 Robles R, Figueras J, Turrión VS. et al. Spanish experience in liver transplantation for hilar and peripheral cholangiocarcinoma. Ann Surg 2004; 239 (02) 265-271
  • 29 Seehofer D, Thelen A, Neumann UP. et al. Extended bile duct resection and [corrected] liver and transplantation in patients with hilar cholangiocarcinoma: long-term results. Liver Transpl 2009; 15 (11) 1499-1507
  • 30 Sapisochin G, de Sevilla EF, Echeverri J, Charco R. Management of “very early” hepatocellular carcinoma on cirrhotic patients. World J Hepatol 2014; 6 (11) 766-775
  • 31 Sapisochin G, Facciuto M, Rubbia-Brandt L. et al; iCCA International Consortium. Liver transplantation for “very early” intrahepatic cholangiocarcinoma: international retrospective study supporting a prospective assessment. Hepatology 2016; 64 (04) 1178-1188
  • 32 Sudan D, DeRoover A, Chinnakotla S. et al. Radiochemotherapy and transplantation allow long-term survival for nonresectable hilar cholangiocarcinoma. Am J Transplant 2002; 2 (08) 774-779
  • 33 De Vreede I, Steers JL, Burch PA. et al. Prolonged disease-free survival after orthotopic liver transplantation plus adjuvant chemoirradiation for cholangiocarcinoma. Liver Transpl 2000; 6 (03) 309-316
  • 34 Heimbach JK, Gores GJ, Haddock MG. et al. Liver transplantation for unresectable perihilar cholangiocarcinoma. Semin Liver Dis 2004; 24 (02) 201-207
  • 35 Darwish Murad S, Kim WR, Harnois DM. et al. Efficacy of neoadjuvant chemoradiation, followed by liver transplantation, for perihilar cholangiocarcinoma at 12 US centers. Gastroenterology 2012; 143 (01) 88-98.e3 , quiz e14
  • 36 Hong JC, Petrowsky H, Kaldas FM. et al. Predictive index for tumor recurrence after liver transplantation for locally advanced intrahepatic and hilar cholangiocarcinoma. J Am Coll Surg 2011; 212 (04) 514-520 , discussion 520–521
  • 37 Lunsford KE, Javle M, Heyne K. et al; Methodist–MD Anderson Joint Cholangiocarcinoma Collaborative Committee (MMAJCCC). Liver transplantation for locally advanced intrahepatic cholangiocarcinoma treated with neoadjuvant therapy: a prospective case-series. Lancet Gastroenterol Hepatol 2018; 3 (05) 337-348
  • 38 McMillan RR, Javle M, Kodali S. et al. Survival following liver transplantation for locally advanced, unresectable intrahepatic cholangiocarcinoma. Am J Transplant 2022; 22 (03) 823-832
  • 39 Ito T, Butler JR, Noguchi D. 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 (03) 386-396
  • 40 Kitajima T, Hibi T, Moonka D, Sapisochin G, Abouljoud MS, Nagai S. Center experience affects liver transplant outcomes in patients with hilar cholangiocarcinoma. Ann Surg Oncol 2020; 27 (13) 5209-5221
  • 41 Eaton JE, Welle CL, Bakhshi Z. et al. Early cholangiocarcinoma detection with magnetic resonance imaging versus ultrasound in primary sclerosing cholangitis. Hepatology 2021; 73 (05) 1868-1881
  • 42 Qin XL, Wang ZR, Shi JS, Lu M, Wang L, He QR. Utility of serum CA19-9 in diagnosis of cholangiocarcinoma: in comparison with CEA. World J Gastroenterol 2004; 10 (03) 427-432
  • 43 Desa LA, Akosa AB, Lazzara S, Domizio P, Krausz T, Benjamin IS. Cytodiagnosis in the management of extrahepatic biliary stricture. Gut 1991; 32 (10) 1188-1191
  • 44 Sugiyama M, Atomi Y, Wada N, Kuroda A, Muto T. Endoscopic transpapillary bile duct biopsy without sphincterotomy for diagnosing biliary strictures: a prospective comparative study with bile and brush cytology. Am J Gastroenterol 1996; 91 (03) 465-467
  • 45 Nanashima A, Yamaguchi H, Nakagoe T. et al. High serum concentrations of sialyl Tn antigen in carcinomas of the biliary tract and pancreas. J Hepatobiliary Pancreat Surg 1999; 6 (04) 391-395
  • 46 Hyman J, Wilczynski SP, Schwarz RE. Extrahepatic bile duct stricture and elevated CA 19-9: malignant or benign?. South Med J 2003; 96 (01) 89-92
  • 47 Chen CY, Shiesh SC, Tsao HC, Lin XZ. The assessment of biliary CA 125, CA 19-9 and CEA in diagnosing cholangiocarcinoma–the influence of sampling time and hepatolithiasis. Hepatogastroenterology 2002; 49 (45) 616-620
  • 48 Kitagawa Y, Iwai M, Muramatsu A. et al. Immunohistochemical localization of CEA, CA19-9 and DU-PAN-2 in hepatitis C virus-infected liver tissues. Histopathology 2002; 40 (05) 472-479
  • 49 Qin X, Shi J, Shi L, Wang Z, Wang L. Clinical value of CA19–9 determination in patients with bile duct carcinoma. Shijie Huaren Xiaohua Zazhi 1999; 7: 814-815
  • 50 Patel AH, Harnois DM, Klee GG, LaRusso NF, Gores GJ. The utility of CA 19-9 in the diagnoses of cholangiocarcinoma in patients without primary sclerosing cholangitis. Am J Gastroenterol 2000; 95 (01) 204-207
  • 51 Gores GJ. Early detection and treatment of cholangiocarcinoma. Liver Transpl 2000; 6 (6, Suppl 2): S30-S34
  • 52 Sapisochin G, Rodríguez de Lope C, Gastaca M. et al “Very early” intrahepatic cholangiocarcinoma in cirrhotic patients: should liver transplantation be reconsidered in these patients?. Am J Transplant 2014; 14 (03) 660-667 DOI: 10.1111/ajt.12591.
  • 53 Iavarone M, Piscaglia F, Vavassori S. et al. Contrast enhanced CT-scan to diagnose intrahepatic cholangiocarcinoma in patients with cirrhosis. J Hepatol 2013; 58 (06) 1188-1193
  • 54 Choi SH, Lee SS, Kim SY. et al. Intrahepatic cholangiocarcinoma in patients with cirrhosis: differentiation from hepatocellular carcinoma by using gadoxetic acid-enhanced MR imaging and dynamic CT. Radiology 2017; 282 (03) 771-781
  • 55 Wildner D, Bernatik T, Greis C, Seitz K, Neurath MF, Strobel D. CEUS in hepatocellular carcinoma and intrahepatic cholangiocellular carcinoma in 320 patients - early or late washout matters: a subanalysis of the DEGUM multicenter trial. Ultraschall Med 2015; 36 (02) 132-139
  • 56 Gatto M, Alvaro D. Cholangiocarcinoma: risk factors and clinical presentation. Eur Rev Med Pharmacol Sci 2010; 14 (04) 363-367
  • 57 Saffioti F, Mavroeidis VK. Review of incidence and outcomes of treatment of cholangiocarcinoma in patients with primary sclerosing cholangitis. World J Gastrointest Oncol 2021; 13 (10) 1336-1366
  • 58 Brandsaeter B, Isoniemi H, Broomé U. et al. Liver transplantation for primary sclerosing cholangitis; predictors and consequences of hepatobiliary malignancy. J Hepatol 2004; 40 (05) 815-822
  • 59 Kim MJ, Choi JY, Chung YE. Evaluation of biliary malignancies using multidetector-row computed tomography. J Comput Assist Tomogr 2010; 34 (04) 496-505
  • 60 Petrowsky H, Wildbrett P, Husarik DB. et al. Impact of integrated positron emission tomography and computed tomography on staging and management of gallbladder cancer and cholangiocarcinoma. J Hepatol 2006; 45 (01) 43-50
  • 61 Lamarca A, Barriuso J, Chander A. et al. 18F-fluorodeoxyglucose positron emission tomography (18FDG-PET) for patients with biliary tract cancer: systematic review and meta-analysis. J Hepatol 2019; 71 (01) 115-129
  • 62 Saluja SS, Sharma R, Pal S, Sahni P, Chattopadhyay TK. Differentiation between benign and malignant hilar obstructions using laboratory and radiological investigations: a prospective study. HPB (Oxford) 2007; 9 (05) 373-382
  • 63 Jhaveri KS, Hosseini-Nik H. MRI of cholangiocarcinoma. J Magn Reson Imaging 2015; 42 (05) 1165-1179
  • 64 Trikudanathan G, Navaneethan U, Njei B, Vargo JJ, Parsi MA. Diagnostic yield of bile duct brushings for cholangiocarcinoma in primary sclerosing cholangitis: a systematic review and meta-analysis. Gastrointest Endosc 2014; 79 (05) 783-789
  • 65 Heimbach JK, Sanchez W, Rosen CB, Gores GJ. Trans-peritoneal fine needle aspiration biopsy of hilar cholangiocarcinoma is associated with disease dissemination. HPB (Oxford) 2011; 13 (05) 356-360
  • 66 Mohamadnejad M, DeWitt JM, Sherman S. et al. Role of EUS for preoperative evaluation of cholangiocarcinoma: a large single-center experience. Gastrointest Endosc 2011; 73 (01) 71-78
  • 67 Polyak A, Kuo A, Sundaram V. Evolution of liver transplant organ allocation policy: current limitations and future directions. World J Hepatol 2021; 13 (08) 830-839
  • 68 Gores GJ, Gish RG, Sudan D, Rosen CB. MELD Exception Study Group. Model for end-stage liver disease (MELD) exception for cholangiocarcinoma or biliary dysplasia. Liver Transpl 2006; 12 (12, Suppl 3): S95-S97
  • 69 Twohig P, Peeraphatdit TB, Mukherjee S. Current status of liver transplantation for cholangiocarcinoma. World J Gastrointest Surg 2022; 14 (01) 1-11
  • 70 OPTN. Organ Procurement and Transplantation Network - OPTN. (n.d.). Accessed March 2, 2024 at: https://optn.transplant.hrsa.gov/media/1200/optn_policies.pdf
  • 71 Yadav S, Xie H, Bin-Riaz I. et al. Neoadjuvant vs. adjuvant chemotherapy for cholangiocarcinoma: a propensity score matched analysis. Eur J Surg Oncol 2019; 45 (08) 1432-1438
  • 72 Welling TH, Feng M, Wan S. et al. Neoadjuvant stereotactic body radiation therapy, capecitabine, and liver transplantation for unresectable hilar cholangiocarcinoma. Liver Transpl 2014; 20 (01) 81-88
  • 73 Duignan S, Maguire D, Ravichand CS. et al. Neoadjuvant chemoradiotherapy followed by liver transplantation for unresectable cholangiocarcinoma: a single-centre national experience. HPB (Oxford) 2014; 16 (01) 91-98
  • 74 Sahai P, Kumar S. External radiotherapy and brachytherapy in the management of extrahepatic and intrahepatic cholangiocarcinoma: available evidence. Br J Radiol 2017; 90 (1076) 20170061
  • 75 Ghali P, Marotta PJ, Yoshida EM. et al. Liver transplantation for incidental cholangiocarcinoma: analysis of the Canadian experience. Liver Transpl 2005; 11 (11) 1412-1416
  • 76 Cambridge WA, Fairfield C, Powell JJ. et al. Meta-analysis and meta-regression of survival after liver transplantation for unresectable perihilar cholangiocarcinoma. Ann Surg 2021; 273 (02) 240-250
  • 77 Lehrke HD, Heimbach JK, Wu TT. et al. Prognostic significance of the histologic response of perihilar cholangiocarcinoma to preoperative neoadjuvant chemoradiation in liver explants. Am J Surg Pathol 2016; 40 (04) 510-518
  • 78 Hazen SMJA, Sluckin TC, Intven MPW. et al. Dutch Snapshot Research Group. Abandonment of routine radiotherapy for nonlocally advanced rectal cancer and oncological outcomes. JAMA Oncol 2024; 10 (02) 202-211
  • 79 Katz MHG, Shi Q, Meyers J. et al. Efficacy of preoperative mFOLFIRINOX vs mFOLFIRINOX plus hypofractionated radiotherapy for borderline resectable adenocarcinoma of the pancreas: the A021501 Phase 2 Randomized Clinical Trial. JAMA Oncol 2022; 8 (09) 1263-1270
  • 80 Pichlmayr R, Weimann A, Tusch G, Schlitt HJ. Indications and role of liver transplantation for malignant tumors. Oncologist 1997; 2 (03) 164-170
  • 81 O'Grady JG, Polson RJ, Rolles K, Calne RY, Williams R. Liver transplantation for malignant disease. Results in 93 consecutive patients. Ann Surg 1988; 207 (04) 373-379
  • 82 Hong JC, Jones CM, Duffy JP. 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 (06) 683-689
  • 83 Sapisochin G, de Lope CR, Gastaca M. et al. Intrahepatic cholangiocarcinoma or mixed hepatocellular-cholangiocarcinoma in patients undergoing liver transplantation: a Spanish matched cohort multicenter study. Ann Surg 2014; 259 (05) 944-952
  • 84 Moris D, Kostakis ID, Machairas N. et al. Comparison between liver transplantation and resection for hilar cholangiocarcinoma: a systematic review and meta-analysis. PLoS One 2019; 14 (07) e0220527
  • 85 Panayotova G, Lunsford KE, Latt NL, Paterno F, Guarrera JV, Pyrsopoulos N. Expanding indications for liver transplantation in the era of liver transplant oncology. World J Gastrointest Surg 2021; 13 (05) 392-405
  • 86 Huang G, Song W, Zhang Y, Yu J, Lv Y, Liu K. Liver transplantation for intrahepatic cholangiocarcinoma: a propensity score-matched analysis. Sci Rep 2023; 13 (01) 10630
  • 87 Sapisochin G, Ivanics T, Heimbach J. Liver transplantation for intrahepatic cholangiocarcinoma: Ready for prime time?. Hepatology 2022; 75 (02) 455-472
  • 88 Lei LL, Song X, Zhao XK. et al. Long-term effect of hospital volume on the postoperative prognosis of 158,618 patients with esophageal squamous cell carcinoma in China. Front Oncol 2023; 12: 1056086
  • 89 Hong TS, Wo JY, Yeap BY. 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 (05) 460-468
  • 90 Takahashi K, Obeid J, Burmeister CS. et al. Intrahepatic cholangiocarcinoma in the liver explant after liver transplantation: histological differentiation and prognosis. Ann Transplant 2016; 21: 208-215
  • 91 De Martin E, Rayar M, Golse N. 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 (06) 785-798
  • 92 Hu XX, Yan LN. Retrospective analysis of prognostic factors after liver transplantation for intrahepatic cholangiocarcinoma in China: a single-center experience. Hepatogastroenterology 2011; 58 (109) 1255-1259
  • 93 Lamarca A, Ross P, Wasan HS. et al. Advanced intrahepatic cholangiocarcinoma: post hoc analysis of the ABC-01, -02, and -03 clinical trials. J Natl Cancer Inst 2020; 112 (02) 200-210
  • 94 Abdelrahim M, Al-Rawi H, Esmail A. et al. Gemcitabine and cisplatin as neo-adjuvant for cholangiocarcinoma patients prior to liver transplantation: case-series. Curr Oncol 2022; 29 (05) 3585-3594
  • 95 Borakati A, Froghi F, Bhogal RH, Mavroeidis VK. Liver transplantation in the management of cholangiocarcinoma: evolution and contemporary advances. World J Gastroenterol 2023; 29 (13) 1969-1981
  • 96 Borakati A, Froghi F, Bhogal RH, Mavroeidis VK. Stereotactic radiotherapy for intrahepatic cholangiocarcinoma. World J Gastrointest Oncol 2022; 14 (08) 1478-1489
  • 97 Edeline J, Touchefeu Y, Guiu B. et al. Radioembolization plus chemotherapy for first-line treatment of locally advanced intrahepatic cholangiocarcinoma: a phase 2 clinical trial. JAMA Oncol 2020; 6 (01) 51-59
  • 98 Ahmed O, Yu Q, Patel M. et al. Yttrium-90 radioembolization and concomitant systemic gemcitabine, cisplatin, and capecitabine as the first-line therapy for locally advanced intrahepatic cholangiocarcinoma. J Vasc Interv Radiol 2023; 34 (04) 702-709
  • 99 Rayar M, Sulpice L, Edeline J. et al. Intra-arterial yttrium-90 radioembolization combined with systemic chemotherapy is a promising method for downstaging unresectable huge intrahepatic cholangiocarcinoma to surgical treatment. Ann Surg Oncol 2015; 22 (09) 3102-3108
  • 100 Villalobos A, Wagstaff W, Guo M. et al. Predictors of successful yttrium-90 radioembolization bridging or downstaging in patients with hepatocellular carcinoma. Can J Gastroenterol Hepatol 2021; 2021: 9926704
  • 101 Sarwar A, Ali A, Ljuboja D. et al. Neoadjuvant yttrium-90 transarterial radioembolization with resin microspheres prescribed using the medical internal radiation dose model for intrahepatic cholangiocarcinoma. J Vasc Interv Radiol 2021; 32 (11) 1560-1568
  • 102 Schartz DA, Porter M, Schartz E. et al. Transarterial yttrium-90 radioembolization for unresectable intrahepatic cholangiocarcinoma: a systematic review and meta-analysis. J Vasc Interv Radiol 2022; 33 (06) 679-686
  • 103 Gupta AN, Gordon AC, Gabr A. et al. Yttrium-90 radioembolization of unresectable intrahepatic cholangiocarcinoma: long-term follow-up for a 136-patient cohort. Cardiovasc Intervent Radiol 2022; 45 (08) 1117-1128
  • 104 Lamarca A, Palmer DH, Wasan HS. et al; Advanced Biliary Cancer Working Group. Second-line FOLFOX chemotherapy versus active symptom control for advanced biliary tract cancer (ABC-06): a phase 3, open-label, randomised, controlled trial. Lancet Oncol 2021; 22 (05) 690-701
  • 105 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 (06) 522-532
  • 106 Kodali S, Connor AA, Thabet S, Brombosz EW, Ghobrial RM. Comparison of resection versus liver transplantation for intrahepatic cholangiocarcinoma: past, present, and future directions. Hepatobiliary Pancreat Dis Int 2024; 23 (02) 129-138
  • 107 Ren A, Li Z, Zhang X, Deng R, Ma Y. A model for predicting post-liver transplantation recurrence in intrahepatic cholangiocarcinoma recipients. J Gastrointest Oncol 2020; 11 (06) 1283-1290
  • 108 Facciuto ME, Singh MK, Lubezky N. et al. Tumors with intrahepatic bile duct differentiation in cirrhosis: implications on outcomes after liver transplantation. Transplantation 2015; 99 (01) 151-157
  • 109 Ziogas IA, Giannis D, Economopoulos KP. et al. Liver transplantation for intrahepatic cholangiocarcinoma: a meta-analysis and meta-regression of survival rates. Transplantation 2021; 105 (10) 2263-2271
  • 110 Krasnodębski M, Grąt M, Wierzchowski M. et al. Analysis of patients with incidental perihilar cholangiocarcinoma: an old and a persistent burden for liver transplantation. Transplant Proc 2020; 52 (08) 2507-2511
  • 111 Cillo U, Fondevila C, Donadon M. et al. Surgery for cholangiocarcinoma. Liver Int 2019; 39 (Suppl, Suppl 1): 143-155
  • 112 Gruttadauria S, Barbera F, Pagano D. et al. Liver transplantation for unresectable intrahepatic cholangiocarcinoma: the role of sequencing genetic profiling. Cancers (Basel) 2021; 13 (23) 6049
  • 113 Lin Y, Peng L, Dong L. et al. Geospatial immune heterogeneity reflects the diverse tumor-immune interactions in intrahepatic cholangiocarcinoma. Cancer Discov 2022; 12 (10) 2350-2371
  • 114 Chen S, Xie Y, Cai Y. et al. Multiomic analysis reveals comprehensive tumor heterogeneity and distinct immune subtypes in multifocal intrahepatic cholangiocarcinoma. Clin Cancer Res 2022; 28 (09) 1896-1910
  • 115 Dong LQ, Shi Y, Ma LJ. et al. Spatial and temporal clonal evolution of intrahepatic cholangiocarcinoma. J Hepatol 2018; 69 (01) 89-98
  • 116 Walter D, Döring C, Feldhahn M. et al. Intratumoral heterogeneity of intrahepatic cholangiocarcinoma. Oncotarget 2017; 8 (09) 14957-14968
  • 117 Clavien PA, Lesurtel M, Bossuyt PM, Gores GJ, Langer B, Perrier A. OLT for HCC Consensus Group. Recommendations for liver transplantation for hepatocellular carcinoma: an international consensus conference report. Lancet Oncol 2012; 13 (01) e11-e22
  • 118 Zhang XF, Chen Q, Kimbrough CW. et al. Lymphadenectomy for intrahepatic cholangiocarcinoma: has nodal evaluation been increasingly adopted by surgeons over time? A national database analysis. J Gastrointest Surg 2018; 22 (04) 668-675
  • 119 Navarro JG, Lee JH, Kang I. et al. Prognostic significance of and risk prediction model for lymph node metastasis in resectable intrahepatic cholangiocarcinoma: do all require lymph node dissection?. HPB (Oxford) 2020; 22 (10) 1411-1419
  • 120 Sposito C, Droz Dit Busset M, Virdis M. et al. The role of lymphadenectomy in the surgical treatment of intrahepatic cholangiocarcinoma: a review. Eur J Surg Oncol 2022; 48 (01) 150-159
  • 121 Li F, Jiang Y, Jiang L. et al. Effect of lymph node resection on prognosis of resectable intrahepatic cholangiocarcinoma: a systematic review and meta-analysis. Front Oncol 2022; 12: 957792
  • 122 Ma KW, Cheung TT, She WH. et al. Diagnostic and prognostic role of 18-FDG PET/CT in the management of resectable biliary tract cancer. World J Surg 2018; 42 (03) 823-834
  • 123 Brierley JD, Gospodarowicz MK, Wittekind C. TNM Classification of Malignant Tumours. John Wiley & Sons; 2017
  • 124 Bagante F, Gani F, Spolverato G. et al. Intrahepatic cholangiocarcinoma: prognosis of patients who did not undergo lymphadenectomy. J Am Coll Surg 2015; 221 (06) 1031-40.e1 , 4
  • 125 Shimada M, Yamashita Y, Aishima S, Shirabe K, Takenaka K, Sugimachi K. Value of lymph node dissection during resection of intrahepatic cholangiocarcinoma. Br J Surg 2001; 88 (11) 1463-1466
  • 126 Bagante F, Spolverato G, Weiss M. et al. Assessment of the lymph node status in patients undergoing liver resection for intrahepatic cholangiocarcinoma: the New Eighth Edition AJCC Staging System. J Gastrointest Surg 2018; 22 (01) 52-59
  • 127 Kim SH, Han DH, Choi GH, Choi JS, Kim KS. Extent of lymph node dissection for accurate staging in intrahepatic cholangiocarcinoma. J Gastrointest Surg 2022; 26 (01) 70-76
  • 128 Sposito C, Ratti F, Cucchetti A. et al. Survival benefit of adequate lymphadenectomy in patients undergoing liver resection for clinically node-negative intrahepatic cholangiocarcinoma. J Hepatol 2023; 78 (02) 356-363
  • 129 Li J, Zhou MH, Ma WJ, Li FY, Deng YL. Extended lymphadenectomy in hilar cholangiocarcinoma: What it will bring?. World J Gastroenterol 2020; 26 (24) 3318-3325
  • 130 Giuliante F, Ardito F, Guglielmi A. et al. Association of lymph node status with survival in patients after liver resection for hilar cholangiocarcinoma in an Italian multicenter analysis. JAMA Surg 2016; 151 (10) 916-922
  • 131 Conci S, Ruzzenente A, Sandri M. et al. What is the most accurate lymph node staging method for perihilar cholangiocarcinoma? Comparison of UICC/AJCC pN stage, number of metastatic lymph nodes, lymph node ratio, and log odds of metastatic lymph nodes. Eur J Surg Oncol 2017; 43 (04) 743-750
  • 132 Kitagawa Y, Nagino M, Kamiya J. et al. Lymph node metastasis from hilar cholangiocarcinoma: audit of 110 patients who underwent regional and paraaortic node dissection. Ann Surg 2001; 233 (03) 385-392
  • 133 Bagante F, Tran T, Spolverato G. et al. Perihilar cholangiocarcinoma: number of nodes examined and optimal lymph node prognostic scheme. J Am Coll Surg 2016; 222 (05) 750-759.e2
  • 134 Mantel HTJ, Wiggers JK, Verheij J. et al. Lymph node micrometastases are associated with worse survival in patients with otherwise node-negative hilar cholangiocarcinoma. Ann Surg Oncol 2015; 22 (Suppl. 03) S1107-S1115
  • 135 Chun YS, Pawlik TM, Vauthey JN. 8th Edition of the AJCC cancer staging manual: pancreas and hepatobiliary cancers. Ann Surg Oncol 2018; 25 (04) 845-847
  • 136 Miyazaki M, Yoshitomi H, Miyakawa S. et al. Clinical practice guidelines for the management of biliary tract cancers 2015: the 2nd English edition. J Hepatobiliary Pancreat Sci 2015; 22 (04) 249-273
  • 137 Ogura Y, Kawarada Y. Surgical strategies for carcinoma of the hepatic duct confluence. Br J Surg 1998; 85 (01) 20-24
  • 138 Miyazaki M, Ito H, Nakagawa K. et al. Parenchyma-preserving hepatectomy in the surgical treatment of hilar cholangiocarcinoma. J Am Coll Surg 1999; 189 (06) 575-583
  • 139 Aoba T, Ebata T, Yokoyama Y. et al. Assessment of nodal status for perihilar cholangiocarcinoma: location, number, or ratio of involved nodes. Ann Surg 2013; 257 (04) 718-725
  • 140 Guglielmi A, Ruzzenente A, Campagnaro T. et al. Patterns and prognostic significance of lymph node dissection for surgical treatment of perihilar and intrahepatic cholangiocarcinoma. J Gastrointest Surg 2013; 17 (11) 1917-1928
  • 141 Ocuin LM, Bağci P, Fisher SB. et al. Discordance between conventional and detailed lymph node analysis in resected biliary carcinoma at or above the cystic duct: are we understaging patients?. Ann Surg Oncol 2013; 20 (13) 4298-4304
  • 142 Oshiro Y, Sasaki R, Kobayashi A. et al. Prognostic relevance of the lymph node ratio in surgical patients with extrahepatic cholangiocarcinoma. Eur J Surg Oncol 2011; 37 (01) 60-64
  • 143 de Jong MC, Marques H, Clary BM. et al. The impact of portal vein resection on outcomes for hilar cholangiocarcinoma: a multi-institutional analysis of 305 cases. Cancer 2012; 118 (19) 4737-4747
  • 144 Hakeem AR, Marangoni G, Chapman SJ. et al. Does the extent of lymphadenectomy, number of lymph nodes, positive lymph node ratio and neutrophil-lymphocyte ratio impact surgical outcome of perihilar cholangiocarcinoma?. Eur J Gastroenterol Hepatol 2014; 26 (09) 1047-1054
  • 145 Murakami Y, Uemura K, Sudo T. et al. Is para-aortic lymph node metastasis a contraindication for radical resection in biliary carcinoma?. World J Surg 2011; 35 (05) 1085-1093
  • 146 Ma WJ, Wu ZR, Hu HJ. et al. Extended lymphadenectomy versus regional lymphadenectomy in resectable hilar cholangiocarcinoma. J Gastrointest Surg 2020; 24 (07) 1619-1629
  • 147 Mao K, Liu J, Sun J. et al. Patterns and prognostic value of lymph node dissection for resected perihilar cholangiocarcinoma. J Gastroenterol Hepatol 2016; 31 (02) 417-426
  • 148 Pandanaboyana S, Bell R, Bartlett AJ, McCall J, Hidalgo E. Meta-analysis of Duct-to-duct versus Roux-en-Y biliary reconstruction following liver transplantation for primary sclerosing cholangitis. Transpl Int 2015; 28 (04) 485-491
  • 149 Kasahara M, Egawa H, Takada Y. et al. Biliary reconstruction in right lobe living-donor liver transplantation: Comparison of different techniques in 321 recipients. Ann Surg 2006; 243 (04) 559-566
  • 150 Safarpour AR, Askari H, Ejtehadi F. et al. Cholangiocarcinoma and liver transplantation: What we know so far?. World J Gastrointest Pathophysiol 2021; 12 (05) 84-105
  • 151 Croome KP, Rosen CB, Heimbach JK, Nagorney DM. Is liver transplantation appropriate for patients with potentially resectable de novo hilar cholangiocarcinoma?. J Am Coll Surg 2015; 221 (01) 130-139
  • 152 Reichman TW, Katchman H, Tanaka T. et al. Living donor versus deceased donor liver transplantation: a surgeon-matched comparison of recipient morbidity and outcomes. Transpl Int 2013; 26 (08) 780-787
  • 153 Callaghan CJ, Charman SC, Muiesan P, Powell JJ, Gimson AE, van der Meulen JHP. UK Liver Transplant Audit. Outcomes of transplantation of livers from donation after circulatory death donors in the UK: a cohort study. BMJ Open 2013; 3 (09) e003287
  • 154 Nasralla D, Coussios CC, Mergental H. et al; Consortium for Organ Preservation in Europe. A randomized trial of normothermic preservation in liver transplantation. Nature 2018; 557 (7703) 50-56
  • 155 2023 Policy Report of the U.S. Organ Procurement and Transplantation Network and the Scientific Registry of Transplant Recipients: Transplant Data 1994-2022. Department of Health and Human Services, Health Resources and Services Administration, Healthcare Systems Bureau, Division of Transplantation, Rockville, MD; United Network for Organ Sharing, Richmond, VA; University Renal Research and Education Association, Ann Arbor, MI. Available at: https://optn.transplant.hrsa.gov/media/eavh5bf3/optn_policies.pdf . Accessed July 24, 2024
  • 156 Bowlus CL, Arrivé L, Bergquist A. et al AASLD practice guidance on primary sclerosing cholangitis and cholangiocarcinoma. Hepatology 2023; 77 (02) 659-702 DOI: 10.1002/hep.32771.
  • 157 Primrose JN, Fox RP, Palmer DH. et al; BILCAP Study Group. Capecitabine compared with observation in resected biliary tract cancer (BILCAP): a randomised, controlled, multicentre, phase 3 study. Lancet Oncol 2019; 20 (05) 663-673
  • 158 Bridgewater J, Fletcher P, Palmer DH. et al; BILCAP Study Group. Long-term outcomes and exploratory analyses of the randomized phase III BILCAP study. J Clin Oncol 2022; 40 (18) 2048-2057
  • 159 Horgan AM, Amir E, Walter T, Knox JJ. Adjuvant therapy in the treatment of biliary tract cancer: a systematic review and meta-analysis. J Clin Oncol 2012; 30 (16) 1934-1940
  • 160 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 (24) 2617-2622
  • 161 Jeong H, Kim KP, Jeong JH. et al. Adjuvant gemcitabine plus cisplatin versus capecitabine in node-positive extrahepatic cholangiocarcinoma: the STAMP randomized trial. Hepatology 2023; 77 (05) 1540-1549
  • 162 Waseem D, Tushar P. Intrahepatic, perihilar and distal cholangiocarcinoma: management and outcomes. Ann Hepatol 2017; 16 (01) 133-139
  • 163 Kendall T, Verheij J, Gaudio E. et al. Anatomical, histomorphological and molecular classification of cholangiocarcinoma. Liver Int 2019; 39 (Suppl. 01) 7-18
  • 164 Rizzo A, Carloni R, Frega G. et al. Intensive follow-up program and oncological outcomes of biliary tract cancer patients after curative-intent surgery: a twenty-year experience in a single tertiary medical center. Curr Oncol 2022; 29 (07) 5084-5090
  • 165 Lamarca A, McNamara MG, Hubner R, Valle JW. Role of ctDNA to predict risk of recurrence following potentially curative resection of biliary tract and pancreatic malignancies. J Clin Oncol 2021; 39 (3, Suppl): 336
  • 166 Boerner T, Drill E, Pak LM. et al. Genetic determinants of outcome in intrahepatic cholangiocarcinoma. Hepatology 2021; 74 (03) 1429-1444
  • 167 Mirallas O, López-Valbuena D, García-Illescas D. et al. Advances in the systemic treatment of therapeutic approaches in biliary tract cancer. ESMO Open 2022; 7 (03) 100503
  • 168 Assistance Publique - Hôpitaux de Paris. Randomized Prospective Multicentric Study: Radio-Chemotherapy and Liver Transplantation Versus Liver Resection to Treat Respectable Hilar Cholangiocarcinoma. clinicaltrials.gov; 2021. Accessed January 1, 2024 at: https://clinicaltrials.gov/study/NCT02232932
  • 169 Gül-Klein S, Schmitz P, Schöning W. et al. The role of immunosuppression for recurrent cholangiocellular carcinoma after liver transplantation. Cancers (Basel) 2022; 14 (12) 2890
  • 170 Resch T, Esser H, Cardini B, Schaefer B, Zoller H, Schneeberger S. Liver transplantation for hilar cholangiocarcinoma (h-CCA): is it the right time?. Transl Gastroenterol Hepatol 2018; 3: 38
  • 171 Ahmad MU, Hanna A, Mohamed AZ. et al. A systematic review of opt-out versus opt-in consent on deceased organ donation and transplantation (2006-2016). World J Surg 2019; 43 (12) 3161-3171
  • 172 Gringeri E, Gambato M, Sapisochin G. et al. Cholangiocarcinoma as an indication for liver transplantation in the era of transplant oncology. J Clin Med 2020; 9 (05) 1353
  • 173 Jarnagin WR, Ruo L, Little SA. et al. Patterns of initial disease recurrence after resection of gallbladder carcinoma and hilar cholangiocarcinoma: implications for adjuvant therapeutic strategies. Cancer 2003; 98 (08) 1689-1700
  • 174 Soares KC, Kamel I, Cosgrove DP, Herman JM, Pawlik TM. Hilar cholangiocarcinoma: diagnosis, treatment options, and management. Hepatobiliary Surg Nutr 2014; 3 (01) 18-34
  • 175 Kim BH, Kim K, Chie EK. et al. Long-term outcome of distal cholangiocarcinoma after pancreaticoduodenectomy followed by adjuvant chemoradiotherapy: a 15-year experience in a single institution. Cancer Res Treat 2017; 49 (02) 473-483
  • 176 Tjaden C, Hinz U, Klaiber U. et al. Distal bile duct cancer: radical (R0 > 1 mm) resection achieves favorable survival. Ann Surg 2023; 277 (01) e112-e118

Address for correspondence

Rafik Mark Ghobrial, MD, PhD, FACS, FRCS(Ed)
Department of Surgery, Houston Methodist Hospital
6550 Fannin Street, Suite 1601 Houston, TX 77030

Publication History

Received: 14 March 2024

Accepted: 29 April 2024

Article published online:
31 July 2024

© 2024. Thieme. All rights reserved.

Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

  • References

  • 1 Loeuillard E, Conboy CB, Gores GJ, Rizvi S. Immunobiology of cholangiocarcinoma. JHEP Rep Innov Hepatol 2019; 1 (04) 297-311
  • 2 Rizvi S, Khan SA, Hallemeier CL, Kelley RK, Gores GJ. Cholangiocarcinoma - evolving concepts and therapeutic strategies. Nat Rev Clin Oncol 2018; 15 (02) 95-111
  • 3 Connor AA, Kodali S, Abdelrahim M, Javle MM, Brombosz EW, Ghobrial RM. Intrahepatic cholangiocarcinoma: the role of liver transplantation, adjunctive treatments, and prognostic biomarkers. Front Oncol 2022; 12: 996710
  • 4 Rizvi S, Gores GJ. Pathogenesis, diagnosis, and management of cholangiocarcinoma. Gastroenterology 2013; 145 (06) 1215-1229
  • 5 Clements O, Eliahoo J, Kim JU, Taylor-Robinson SD, Khan SA. Risk factors for intrahepatic and extrahepatic cholangiocarcinoma: a systematic review and meta-analysis. J Hepatol 2020; 72 (01) 95-103
  • 6 Tyson GL, El-Serag HB. Risk factors for cholangiocarcinoma. Hepatology 2011; 54 (01) 173-184
  • 7 Abdelrahim M, Esmail A, Abudayyeh A. et al. Transplant oncology: an evolving field in cancer care. Cancers (Basel) 2021; 13 (19) 4911
  • 8 Banales JM, Marin JJG, Lamarca A. et al. Cholangiocarcinoma 2020: the next horizon in mechanisms and management. Nat Rev Gastroenterol Hepatol 2020; 17 (09) 557-588
  • 9 Weber SM, Ribero D, O'Reilly EM, Kokudo N, Miyazaki M, Pawlik TM. Intrahepatic cholangiocarcinoma: expert consensus statement. HPB (Oxford) 2015; 17 (08) 669-680
  • 10 DeOliveira ML, Cunningham SC, Cameron JL. et al. Cholangiocarcinoma: thirty-one-year experience with 564 patients at a single institution. Ann Surg 2007; 245 (05) 755-762
  • 11 Jarnagin WR, Fong Y, DeMatteo RP. et al. Staging, resectability, and outcome in 225 patients with hilar cholangiocarcinoma. Ann Surg 2001; 234 (04) 507-517 , discussion 517–519
  • 12 Kobayashi A, Miwa S, Nakata T, Miyagawa S. Disease recurrence patterns after R0 resection of hilar cholangiocarcinoma. Br J Surg 2010; 97 (01) 56-64
  • 13 Kosuge T, Yamamoto J, Shimada K, Yamasaki S, Makuuchi M. Improved surgical results for hilar cholangiocarcinoma with procedures including major hepatic resection. Ann Surg 1999; 230 (05) 663-671
  • 14 Rea DJ, Heimbach JK, Rosen CB. et al. Liver transplantation with neoadjuvant chemoradiation is more effective than resection for hilar cholangiocarcinoma. Ann Surg 2005; 242 (03) 451-458 , discussion 458–461
  • 15 Rea DJ, Munoz-Juarez M, Farnell MB. et al. Major hepatic resection for hilar cholangiocarcinoma: analysis of 46 patients. Arch Surg 2004; 139 (05) 514-523 , discussion 523–525
  • 16 Su CH, Tsay SH, Wu CC. et al. Factors influencing postoperative morbidity, mortality, and survival after resection for hilar cholangiocarcinoma. Ann Surg 1996; 223 (04) 384-394
  • 17 Washburn WK, Lewis WD, Jenkins RL. Aggressive surgical resection for cholangiocarcinoma. Arch Surg 1995; 130 (03) 270-276
  • 18 Spolverato G, Bagante F, Tsilimigras D, Ejaz A, Cloyd J, Pawlik TM. Management and outcomes among patients with mixed hepatocholangiocellular carcinoma: a population-based analysis. J Surg Oncol 2019; 119 (03) 278-287
  • 19 Brindley PJ, Bachini M, Ilyas SI. et al. Cholangiocarcinoma. Nat Rev Dis Primers 2021; 7 (01) 65
  • 20 Tomlinson JL, Valle JW, Ilyas SI. Immunobiology of cholangiocarcinoma. J Hepatol 2023; 79 (03) 867-875
  • 21 Lee H, Ross JS. The potential role of comprehensive genomic profiling to guide targeted therapy for patients with biliary cancer. Therap Adv Gastroenterol 2017; 10 (06) 507-520
  • 22 Mazzaferro V, Gorgen A, Roayaie S, Droz Dit Busset M, Sapisochin G. Liver resection and transplantation for intrahepatic cholangiocarcinoma. J Hepatol 2020; 72 (02) 364-377
  • 23 Hyder O, Marques H, Pulitano C. et al. A nomogram to predict long-term survival after resection for intrahepatic cholangiocarcinoma: an Eastern and Western experience. JAMA Surg 2014; 149 (05) 432-438
  • 24 Meza-Junco J, Montano-Loza AJ, Ma M, Wong W, Sawyer MB, Bain VG. Cholangiocarcinoma: has there been any progress?. Can J Gastroenterol 2010; 24 (01) 52-57
  • 25 Casavilla FA, Marsh JW, Iwatsuki S. et al. Hepatic resection and transplantation for peripheral cholangiocarcinoma. J Am Coll Surg 1997; 185 (05) 429-436
  • 26 Meyer CG, Penn I, James L. Liver transplantation for cholangiocarcinoma: results in 207 patients. Transplantation 2000; 69 (08) 1633-1637
  • 27 Shimoda M, Farmer DG, Colquhoun SD. et al. Liver transplantation for cholangiocellular carcinoma: analysis of a single-center experience and review of the literature. Liver Transpl 2001; 7 (12) 1023-1033
  • 28 Robles R, Figueras J, Turrión VS. et al. Spanish experience in liver transplantation for hilar and peripheral cholangiocarcinoma. Ann Surg 2004; 239 (02) 265-271
  • 29 Seehofer D, Thelen A, Neumann UP. et al. Extended bile duct resection and [corrected] liver and transplantation in patients with hilar cholangiocarcinoma: long-term results. Liver Transpl 2009; 15 (11) 1499-1507
  • 30 Sapisochin G, de Sevilla EF, Echeverri J, Charco R. Management of “very early” hepatocellular carcinoma on cirrhotic patients. World J Hepatol 2014; 6 (11) 766-775
  • 31 Sapisochin G, Facciuto M, Rubbia-Brandt L. et al; iCCA International Consortium. Liver transplantation for “very early” intrahepatic cholangiocarcinoma: international retrospective study supporting a prospective assessment. Hepatology 2016; 64 (04) 1178-1188
  • 32 Sudan D, DeRoover A, Chinnakotla S. et al. Radiochemotherapy and transplantation allow long-term survival for nonresectable hilar cholangiocarcinoma. Am J Transplant 2002; 2 (08) 774-779
  • 33 De Vreede I, Steers JL, Burch PA. et al. Prolonged disease-free survival after orthotopic liver transplantation plus adjuvant chemoirradiation for cholangiocarcinoma. Liver Transpl 2000; 6 (03) 309-316
  • 34 Heimbach JK, Gores GJ, Haddock MG. et al. Liver transplantation for unresectable perihilar cholangiocarcinoma. Semin Liver Dis 2004; 24 (02) 201-207
  • 35 Darwish Murad S, Kim WR, Harnois DM. et al. Efficacy of neoadjuvant chemoradiation, followed by liver transplantation, for perihilar cholangiocarcinoma at 12 US centers. Gastroenterology 2012; 143 (01) 88-98.e3 , quiz e14
  • 36 Hong JC, Petrowsky H, Kaldas FM. et al. Predictive index for tumor recurrence after liver transplantation for locally advanced intrahepatic and hilar cholangiocarcinoma. J Am Coll Surg 2011; 212 (04) 514-520 , discussion 520–521
  • 37 Lunsford KE, Javle M, Heyne K. et al; Methodist–MD Anderson Joint Cholangiocarcinoma Collaborative Committee (MMAJCCC). Liver transplantation for locally advanced intrahepatic cholangiocarcinoma treated with neoadjuvant therapy: a prospective case-series. Lancet Gastroenterol Hepatol 2018; 3 (05) 337-348
  • 38 McMillan RR, Javle M, Kodali S. et al. Survival following liver transplantation for locally advanced, unresectable intrahepatic cholangiocarcinoma. Am J Transplant 2022; 22 (03) 823-832
  • 39 Ito T, Butler JR, Noguchi D. 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 (03) 386-396
  • 40 Kitajima T, Hibi T, Moonka D, Sapisochin G, Abouljoud MS, Nagai S. Center experience affects liver transplant outcomes in patients with hilar cholangiocarcinoma. Ann Surg Oncol 2020; 27 (13) 5209-5221
  • 41 Eaton JE, Welle CL, Bakhshi Z. et al. Early cholangiocarcinoma detection with magnetic resonance imaging versus ultrasound in primary sclerosing cholangitis. Hepatology 2021; 73 (05) 1868-1881
  • 42 Qin XL, Wang ZR, Shi JS, Lu M, Wang L, He QR. Utility of serum CA19-9 in diagnosis of cholangiocarcinoma: in comparison with CEA. World J Gastroenterol 2004; 10 (03) 427-432
  • 43 Desa LA, Akosa AB, Lazzara S, Domizio P, Krausz T, Benjamin IS. Cytodiagnosis in the management of extrahepatic biliary stricture. Gut 1991; 32 (10) 1188-1191
  • 44 Sugiyama M, Atomi Y, Wada N, Kuroda A, Muto T. Endoscopic transpapillary bile duct biopsy without sphincterotomy for diagnosing biliary strictures: a prospective comparative study with bile and brush cytology. Am J Gastroenterol 1996; 91 (03) 465-467
  • 45 Nanashima A, Yamaguchi H, Nakagoe T. et al. High serum concentrations of sialyl Tn antigen in carcinomas of the biliary tract and pancreas. J Hepatobiliary Pancreat Surg 1999; 6 (04) 391-395
  • 46 Hyman J, Wilczynski SP, Schwarz RE. Extrahepatic bile duct stricture and elevated CA 19-9: malignant or benign?. South Med J 2003; 96 (01) 89-92
  • 47 Chen CY, Shiesh SC, Tsao HC, Lin XZ. The assessment of biliary CA 125, CA 19-9 and CEA in diagnosing cholangiocarcinoma–the influence of sampling time and hepatolithiasis. Hepatogastroenterology 2002; 49 (45) 616-620
  • 48 Kitagawa Y, Iwai M, Muramatsu A. et al. Immunohistochemical localization of CEA, CA19-9 and DU-PAN-2 in hepatitis C virus-infected liver tissues. Histopathology 2002; 40 (05) 472-479
  • 49 Qin X, Shi J, Shi L, Wang Z, Wang L. Clinical value of CA19–9 determination in patients with bile duct carcinoma. Shijie Huaren Xiaohua Zazhi 1999; 7: 814-815
  • 50 Patel AH, Harnois DM, Klee GG, LaRusso NF, Gores GJ. The utility of CA 19-9 in the diagnoses of cholangiocarcinoma in patients without primary sclerosing cholangitis. Am J Gastroenterol 2000; 95 (01) 204-207
  • 51 Gores GJ. Early detection and treatment of cholangiocarcinoma. Liver Transpl 2000; 6 (6, Suppl 2): S30-S34
  • 52 Sapisochin G, Rodríguez de Lope C, Gastaca M. et al “Very early” intrahepatic cholangiocarcinoma in cirrhotic patients: should liver transplantation be reconsidered in these patients?. Am J Transplant 2014; 14 (03) 660-667 DOI: 10.1111/ajt.12591.
  • 53 Iavarone M, Piscaglia F, Vavassori S. et al. Contrast enhanced CT-scan to diagnose intrahepatic cholangiocarcinoma in patients with cirrhosis. J Hepatol 2013; 58 (06) 1188-1193
  • 54 Choi SH, Lee SS, Kim SY. et al. Intrahepatic cholangiocarcinoma in patients with cirrhosis: differentiation from hepatocellular carcinoma by using gadoxetic acid-enhanced MR imaging and dynamic CT. Radiology 2017; 282 (03) 771-781
  • 55 Wildner D, Bernatik T, Greis C, Seitz K, Neurath MF, Strobel D. CEUS in hepatocellular carcinoma and intrahepatic cholangiocellular carcinoma in 320 patients - early or late washout matters: a subanalysis of the DEGUM multicenter trial. Ultraschall Med 2015; 36 (02) 132-139
  • 56 Gatto M, Alvaro D. Cholangiocarcinoma: risk factors and clinical presentation. Eur Rev Med Pharmacol Sci 2010; 14 (04) 363-367
  • 57 Saffioti F, Mavroeidis VK. Review of incidence and outcomes of treatment of cholangiocarcinoma in patients with primary sclerosing cholangitis. World J Gastrointest Oncol 2021; 13 (10) 1336-1366
  • 58 Brandsaeter B, Isoniemi H, Broomé U. et al. Liver transplantation for primary sclerosing cholangitis; predictors and consequences of hepatobiliary malignancy. J Hepatol 2004; 40 (05) 815-822
  • 59 Kim MJ, Choi JY, Chung YE. Evaluation of biliary malignancies using multidetector-row computed tomography. J Comput Assist Tomogr 2010; 34 (04) 496-505
  • 60 Petrowsky H, Wildbrett P, Husarik DB. et al. Impact of integrated positron emission tomography and computed tomography on staging and management of gallbladder cancer and cholangiocarcinoma. J Hepatol 2006; 45 (01) 43-50
  • 61 Lamarca A, Barriuso J, Chander A. et al. 18F-fluorodeoxyglucose positron emission tomography (18FDG-PET) for patients with biliary tract cancer: systematic review and meta-analysis. J Hepatol 2019; 71 (01) 115-129
  • 62 Saluja SS, Sharma R, Pal S, Sahni P, Chattopadhyay TK. Differentiation between benign and malignant hilar obstructions using laboratory and radiological investigations: a prospective study. HPB (Oxford) 2007; 9 (05) 373-382
  • 63 Jhaveri KS, Hosseini-Nik H. MRI of cholangiocarcinoma. J Magn Reson Imaging 2015; 42 (05) 1165-1179
  • 64 Trikudanathan G, Navaneethan U, Njei B, Vargo JJ, Parsi MA. Diagnostic yield of bile duct brushings for cholangiocarcinoma in primary sclerosing cholangitis: a systematic review and meta-analysis. Gastrointest Endosc 2014; 79 (05) 783-789
  • 65 Heimbach JK, Sanchez W, Rosen CB, Gores GJ. Trans-peritoneal fine needle aspiration biopsy of hilar cholangiocarcinoma is associated with disease dissemination. HPB (Oxford) 2011; 13 (05) 356-360
  • 66 Mohamadnejad M, DeWitt JM, Sherman S. et al. Role of EUS for preoperative evaluation of cholangiocarcinoma: a large single-center experience. Gastrointest Endosc 2011; 73 (01) 71-78
  • 67 Polyak A, Kuo A, Sundaram V. Evolution of liver transplant organ allocation policy: current limitations and future directions. World J Hepatol 2021; 13 (08) 830-839
  • 68 Gores GJ, Gish RG, Sudan D, Rosen CB. MELD Exception Study Group. Model for end-stage liver disease (MELD) exception for cholangiocarcinoma or biliary dysplasia. Liver Transpl 2006; 12 (12, Suppl 3): S95-S97
  • 69 Twohig P, Peeraphatdit TB, Mukherjee S. Current status of liver transplantation for cholangiocarcinoma. World J Gastrointest Surg 2022; 14 (01) 1-11
  • 70 OPTN. Organ Procurement and Transplantation Network - OPTN. (n.d.). Accessed March 2, 2024 at: https://optn.transplant.hrsa.gov/media/1200/optn_policies.pdf
  • 71 Yadav S, Xie H, Bin-Riaz I. et al. Neoadjuvant vs. adjuvant chemotherapy for cholangiocarcinoma: a propensity score matched analysis. Eur J Surg Oncol 2019; 45 (08) 1432-1438
  • 72 Welling TH, Feng M, Wan S. et al. Neoadjuvant stereotactic body radiation therapy, capecitabine, and liver transplantation for unresectable hilar cholangiocarcinoma. Liver Transpl 2014; 20 (01) 81-88
  • 73 Duignan S, Maguire D, Ravichand CS. et al. Neoadjuvant chemoradiotherapy followed by liver transplantation for unresectable cholangiocarcinoma: a single-centre national experience. HPB (Oxford) 2014; 16 (01) 91-98
  • 74 Sahai P, Kumar S. External radiotherapy and brachytherapy in the management of extrahepatic and intrahepatic cholangiocarcinoma: available evidence. Br J Radiol 2017; 90 (1076) 20170061
  • 75 Ghali P, Marotta PJ, Yoshida EM. et al. Liver transplantation for incidental cholangiocarcinoma: analysis of the Canadian experience. Liver Transpl 2005; 11 (11) 1412-1416
  • 76 Cambridge WA, Fairfield C, Powell JJ. et al. Meta-analysis and meta-regression of survival after liver transplantation for unresectable perihilar cholangiocarcinoma. Ann Surg 2021; 273 (02) 240-250
  • 77 Lehrke HD, Heimbach JK, Wu TT. et al. Prognostic significance of the histologic response of perihilar cholangiocarcinoma to preoperative neoadjuvant chemoradiation in liver explants. Am J Surg Pathol 2016; 40 (04) 510-518
  • 78 Hazen SMJA, Sluckin TC, Intven MPW. et al. Dutch Snapshot Research Group. Abandonment of routine radiotherapy for nonlocally advanced rectal cancer and oncological outcomes. JAMA Oncol 2024; 10 (02) 202-211
  • 79 Katz MHG, Shi Q, Meyers J. et al. Efficacy of preoperative mFOLFIRINOX vs mFOLFIRINOX plus hypofractionated radiotherapy for borderline resectable adenocarcinoma of the pancreas: the A021501 Phase 2 Randomized Clinical Trial. JAMA Oncol 2022; 8 (09) 1263-1270
  • 80 Pichlmayr R, Weimann A, Tusch G, Schlitt HJ. Indications and role of liver transplantation for malignant tumors. Oncologist 1997; 2 (03) 164-170
  • 81 O'Grady JG, Polson RJ, Rolles K, Calne RY, Williams R. Liver transplantation for malignant disease. Results in 93 consecutive patients. Ann Surg 1988; 207 (04) 373-379
  • 82 Hong JC, Jones CM, Duffy JP. 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 (06) 683-689
  • 83 Sapisochin G, de Lope CR, Gastaca M. et al. Intrahepatic cholangiocarcinoma or mixed hepatocellular-cholangiocarcinoma in patients undergoing liver transplantation: a Spanish matched cohort multicenter study. Ann Surg 2014; 259 (05) 944-952
  • 84 Moris D, Kostakis ID, Machairas N. et al. Comparison between liver transplantation and resection for hilar cholangiocarcinoma: a systematic review and meta-analysis. PLoS One 2019; 14 (07) e0220527
  • 85 Panayotova G, Lunsford KE, Latt NL, Paterno F, Guarrera JV, Pyrsopoulos N. Expanding indications for liver transplantation in the era of liver transplant oncology. World J Gastrointest Surg 2021; 13 (05) 392-405
  • 86 Huang G, Song W, Zhang Y, Yu J, Lv Y, Liu K. Liver transplantation for intrahepatic cholangiocarcinoma: a propensity score-matched analysis. Sci Rep 2023; 13 (01) 10630
  • 87 Sapisochin G, Ivanics T, Heimbach J. Liver transplantation for intrahepatic cholangiocarcinoma: Ready for prime time?. Hepatology 2022; 75 (02) 455-472
  • 88 Lei LL, Song X, Zhao XK. et al. Long-term effect of hospital volume on the postoperative prognosis of 158,618 patients with esophageal squamous cell carcinoma in China. Front Oncol 2023; 12: 1056086
  • 89 Hong TS, Wo JY, Yeap BY. 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 (05) 460-468
  • 90 Takahashi K, Obeid J, Burmeister CS. et al. Intrahepatic cholangiocarcinoma in the liver explant after liver transplantation: histological differentiation and prognosis. Ann Transplant 2016; 21: 208-215
  • 91 De Martin E, Rayar M, Golse N. 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 (06) 785-798
  • 92 Hu XX, Yan LN. Retrospective analysis of prognostic factors after liver transplantation for intrahepatic cholangiocarcinoma in China: a single-center experience. Hepatogastroenterology 2011; 58 (109) 1255-1259
  • 93 Lamarca A, Ross P, Wasan HS. et al. Advanced intrahepatic cholangiocarcinoma: post hoc analysis of the ABC-01, -02, and -03 clinical trials. J Natl Cancer Inst 2020; 112 (02) 200-210
  • 94 Abdelrahim M, Al-Rawi H, Esmail A. et al. Gemcitabine and cisplatin as neo-adjuvant for cholangiocarcinoma patients prior to liver transplantation: case-series. Curr Oncol 2022; 29 (05) 3585-3594
  • 95 Borakati A, Froghi F, Bhogal RH, Mavroeidis VK. Liver transplantation in the management of cholangiocarcinoma: evolution and contemporary advances. World J Gastroenterol 2023; 29 (13) 1969-1981
  • 96 Borakati A, Froghi F, Bhogal RH, Mavroeidis VK. Stereotactic radiotherapy for intrahepatic cholangiocarcinoma. World J Gastrointest Oncol 2022; 14 (08) 1478-1489
  • 97 Edeline J, Touchefeu Y, Guiu B. et al. Radioembolization plus chemotherapy for first-line treatment of locally advanced intrahepatic cholangiocarcinoma: a phase 2 clinical trial. JAMA Oncol 2020; 6 (01) 51-59
  • 98 Ahmed O, Yu Q, Patel M. et al. Yttrium-90 radioembolization and concomitant systemic gemcitabine, cisplatin, and capecitabine as the first-line therapy for locally advanced intrahepatic cholangiocarcinoma. J Vasc Interv Radiol 2023; 34 (04) 702-709
  • 99 Rayar M, Sulpice L, Edeline J. et al. Intra-arterial yttrium-90 radioembolization combined with systemic chemotherapy is a promising method for downstaging unresectable huge intrahepatic cholangiocarcinoma to surgical treatment. Ann Surg Oncol 2015; 22 (09) 3102-3108
  • 100 Villalobos A, Wagstaff W, Guo M. et al. Predictors of successful yttrium-90 radioembolization bridging or downstaging in patients with hepatocellular carcinoma. Can J Gastroenterol Hepatol 2021; 2021: 9926704
  • 101 Sarwar A, Ali A, Ljuboja D. et al. Neoadjuvant yttrium-90 transarterial radioembolization with resin microspheres prescribed using the medical internal radiation dose model for intrahepatic cholangiocarcinoma. J Vasc Interv Radiol 2021; 32 (11) 1560-1568
  • 102 Schartz DA, Porter M, Schartz E. et al. Transarterial yttrium-90 radioembolization for unresectable intrahepatic cholangiocarcinoma: a systematic review and meta-analysis. J Vasc Interv Radiol 2022; 33 (06) 679-686
  • 103 Gupta AN, Gordon AC, Gabr A. et al. Yttrium-90 radioembolization of unresectable intrahepatic cholangiocarcinoma: long-term follow-up for a 136-patient cohort. Cardiovasc Intervent Radiol 2022; 45 (08) 1117-1128
  • 104 Lamarca A, Palmer DH, Wasan HS. et al; Advanced Biliary Cancer Working Group. Second-line FOLFOX chemotherapy versus active symptom control for advanced biliary tract cancer (ABC-06): a phase 3, open-label, randomised, controlled trial. Lancet Oncol 2021; 22 (05) 690-701
  • 105 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 (06) 522-532
  • 106 Kodali S, Connor AA, Thabet S, Brombosz EW, Ghobrial RM. Comparison of resection versus liver transplantation for intrahepatic cholangiocarcinoma: past, present, and future directions. Hepatobiliary Pancreat Dis Int 2024; 23 (02) 129-138
  • 107 Ren A, Li Z, Zhang X, Deng R, Ma Y. A model for predicting post-liver transplantation recurrence in intrahepatic cholangiocarcinoma recipients. J Gastrointest Oncol 2020; 11 (06) 1283-1290
  • 108 Facciuto ME, Singh MK, Lubezky N. et al. Tumors with intrahepatic bile duct differentiation in cirrhosis: implications on outcomes after liver transplantation. Transplantation 2015; 99 (01) 151-157
  • 109 Ziogas IA, Giannis D, Economopoulos KP. et al. Liver transplantation for intrahepatic cholangiocarcinoma: a meta-analysis and meta-regression of survival rates. Transplantation 2021; 105 (10) 2263-2271
  • 110 Krasnodębski M, Grąt M, Wierzchowski M. et al. Analysis of patients with incidental perihilar cholangiocarcinoma: an old and a persistent burden for liver transplantation. Transplant Proc 2020; 52 (08) 2507-2511
  • 111 Cillo U, Fondevila C, Donadon M. et al. Surgery for cholangiocarcinoma. Liver Int 2019; 39 (Suppl, Suppl 1): 143-155
  • 112 Gruttadauria S, Barbera F, Pagano D. et al. Liver transplantation for unresectable intrahepatic cholangiocarcinoma: the role of sequencing genetic profiling. Cancers (Basel) 2021; 13 (23) 6049
  • 113 Lin Y, Peng L, Dong L. et al. Geospatial immune heterogeneity reflects the diverse tumor-immune interactions in intrahepatic cholangiocarcinoma. Cancer Discov 2022; 12 (10) 2350-2371
  • 114 Chen S, Xie Y, Cai Y. et al. Multiomic analysis reveals comprehensive tumor heterogeneity and distinct immune subtypes in multifocal intrahepatic cholangiocarcinoma. Clin Cancer Res 2022; 28 (09) 1896-1910
  • 115 Dong LQ, Shi Y, Ma LJ. et al. Spatial and temporal clonal evolution of intrahepatic cholangiocarcinoma. J Hepatol 2018; 69 (01) 89-98
  • 116 Walter D, Döring C, Feldhahn M. et al. Intratumoral heterogeneity of intrahepatic cholangiocarcinoma. Oncotarget 2017; 8 (09) 14957-14968
  • 117 Clavien PA, Lesurtel M, Bossuyt PM, Gores GJ, Langer B, Perrier A. OLT for HCC Consensus Group. Recommendations for liver transplantation for hepatocellular carcinoma: an international consensus conference report. Lancet Oncol 2012; 13 (01) e11-e22
  • 118 Zhang XF, Chen Q, Kimbrough CW. et al. Lymphadenectomy for intrahepatic cholangiocarcinoma: has nodal evaluation been increasingly adopted by surgeons over time? A national database analysis. J Gastrointest Surg 2018; 22 (04) 668-675
  • 119 Navarro JG, Lee JH, Kang I. et al. Prognostic significance of and risk prediction model for lymph node metastasis in resectable intrahepatic cholangiocarcinoma: do all require lymph node dissection?. HPB (Oxford) 2020; 22 (10) 1411-1419
  • 120 Sposito C, Droz Dit Busset M, Virdis M. et al. The role of lymphadenectomy in the surgical treatment of intrahepatic cholangiocarcinoma: a review. Eur J Surg Oncol 2022; 48 (01) 150-159
  • 121 Li F, Jiang Y, Jiang L. et al. Effect of lymph node resection on prognosis of resectable intrahepatic cholangiocarcinoma: a systematic review and meta-analysis. Front Oncol 2022; 12: 957792
  • 122 Ma KW, Cheung TT, She WH. et al. Diagnostic and prognostic role of 18-FDG PET/CT in the management of resectable biliary tract cancer. World J Surg 2018; 42 (03) 823-834
  • 123 Brierley JD, Gospodarowicz MK, Wittekind C. TNM Classification of Malignant Tumours. John Wiley & Sons; 2017
  • 124 Bagante F, Gani F, Spolverato G. et al. Intrahepatic cholangiocarcinoma: prognosis of patients who did not undergo lymphadenectomy. J Am Coll Surg 2015; 221 (06) 1031-40.e1 , 4
  • 125 Shimada M, Yamashita Y, Aishima S, Shirabe K, Takenaka K, Sugimachi K. Value of lymph node dissection during resection of intrahepatic cholangiocarcinoma. Br J Surg 2001; 88 (11) 1463-1466
  • 126 Bagante F, Spolverato G, Weiss M. et al. Assessment of the lymph node status in patients undergoing liver resection for intrahepatic cholangiocarcinoma: the New Eighth Edition AJCC Staging System. J Gastrointest Surg 2018; 22 (01) 52-59
  • 127 Kim SH, Han DH, Choi GH, Choi JS, Kim KS. Extent of lymph node dissection for accurate staging in intrahepatic cholangiocarcinoma. J Gastrointest Surg 2022; 26 (01) 70-76
  • 128 Sposito C, Ratti F, Cucchetti A. et al. Survival benefit of adequate lymphadenectomy in patients undergoing liver resection for clinically node-negative intrahepatic cholangiocarcinoma. J Hepatol 2023; 78 (02) 356-363
  • 129 Li J, Zhou MH, Ma WJ, Li FY, Deng YL. Extended lymphadenectomy in hilar cholangiocarcinoma: What it will bring?. World J Gastroenterol 2020; 26 (24) 3318-3325
  • 130 Giuliante F, Ardito F, Guglielmi A. et al. Association of lymph node status with survival in patients after liver resection for hilar cholangiocarcinoma in an Italian multicenter analysis. JAMA Surg 2016; 151 (10) 916-922
  • 131 Conci S, Ruzzenente A, Sandri M. et al. What is the most accurate lymph node staging method for perihilar cholangiocarcinoma? Comparison of UICC/AJCC pN stage, number of metastatic lymph nodes, lymph node ratio, and log odds of metastatic lymph nodes. Eur J Surg Oncol 2017; 43 (04) 743-750
  • 132 Kitagawa Y, Nagino M, Kamiya J. et al. Lymph node metastasis from hilar cholangiocarcinoma: audit of 110 patients who underwent regional and paraaortic node dissection. Ann Surg 2001; 233 (03) 385-392
  • 133 Bagante F, Tran T, Spolverato G. et al. Perihilar cholangiocarcinoma: number of nodes examined and optimal lymph node prognostic scheme. J Am Coll Surg 2016; 222 (05) 750-759.e2
  • 134 Mantel HTJ, Wiggers JK, Verheij J. et al. Lymph node micrometastases are associated with worse survival in patients with otherwise node-negative hilar cholangiocarcinoma. Ann Surg Oncol 2015; 22 (Suppl. 03) S1107-S1115
  • 135 Chun YS, Pawlik TM, Vauthey JN. 8th Edition of the AJCC cancer staging manual: pancreas and hepatobiliary cancers. Ann Surg Oncol 2018; 25 (04) 845-847
  • 136 Miyazaki M, Yoshitomi H, Miyakawa S. et al. Clinical practice guidelines for the management of biliary tract cancers 2015: the 2nd English edition. J Hepatobiliary Pancreat Sci 2015; 22 (04) 249-273
  • 137 Ogura Y, Kawarada Y. Surgical strategies for carcinoma of the hepatic duct confluence. Br J Surg 1998; 85 (01) 20-24
  • 138 Miyazaki M, Ito H, Nakagawa K. et al. Parenchyma-preserving hepatectomy in the surgical treatment of hilar cholangiocarcinoma. J Am Coll Surg 1999; 189 (06) 575-583
  • 139 Aoba T, Ebata T, Yokoyama Y. et al. Assessment of nodal status for perihilar cholangiocarcinoma: location, number, or ratio of involved nodes. Ann Surg 2013; 257 (04) 718-725
  • 140 Guglielmi A, Ruzzenente A, Campagnaro T. et al. Patterns and prognostic significance of lymph node dissection for surgical treatment of perihilar and intrahepatic cholangiocarcinoma. J Gastrointest Surg 2013; 17 (11) 1917-1928
  • 141 Ocuin LM, Bağci P, Fisher SB. et al. Discordance between conventional and detailed lymph node analysis in resected biliary carcinoma at or above the cystic duct: are we understaging patients?. Ann Surg Oncol 2013; 20 (13) 4298-4304
  • 142 Oshiro Y, Sasaki R, Kobayashi A. et al. Prognostic relevance of the lymph node ratio in surgical patients with extrahepatic cholangiocarcinoma. Eur J Surg Oncol 2011; 37 (01) 60-64
  • 143 de Jong MC, Marques H, Clary BM. et al. The impact of portal vein resection on outcomes for hilar cholangiocarcinoma: a multi-institutional analysis of 305 cases. Cancer 2012; 118 (19) 4737-4747
  • 144 Hakeem AR, Marangoni G, Chapman SJ. et al. Does the extent of lymphadenectomy, number of lymph nodes, positive lymph node ratio and neutrophil-lymphocyte ratio impact surgical outcome of perihilar cholangiocarcinoma?. Eur J Gastroenterol Hepatol 2014; 26 (09) 1047-1054
  • 145 Murakami Y, Uemura K, Sudo T. et al. Is para-aortic lymph node metastasis a contraindication for radical resection in biliary carcinoma?. World J Surg 2011; 35 (05) 1085-1093
  • 146 Ma WJ, Wu ZR, Hu HJ. et al. Extended lymphadenectomy versus regional lymphadenectomy in resectable hilar cholangiocarcinoma. J Gastrointest Surg 2020; 24 (07) 1619-1629
  • 147 Mao K, Liu J, Sun J. et al. Patterns and prognostic value of lymph node dissection for resected perihilar cholangiocarcinoma. J Gastroenterol Hepatol 2016; 31 (02) 417-426
  • 148 Pandanaboyana S, Bell R, Bartlett AJ, McCall J, Hidalgo E. Meta-analysis of Duct-to-duct versus Roux-en-Y biliary reconstruction following liver transplantation for primary sclerosing cholangitis. Transpl Int 2015; 28 (04) 485-491
  • 149 Kasahara M, Egawa H, Takada Y. et al. Biliary reconstruction in right lobe living-donor liver transplantation: Comparison of different techniques in 321 recipients. Ann Surg 2006; 243 (04) 559-566
  • 150 Safarpour AR, Askari H, Ejtehadi F. et al. Cholangiocarcinoma and liver transplantation: What we know so far?. World J Gastrointest Pathophysiol 2021; 12 (05) 84-105
  • 151 Croome KP, Rosen CB, Heimbach JK, Nagorney DM. Is liver transplantation appropriate for patients with potentially resectable de novo hilar cholangiocarcinoma?. J Am Coll Surg 2015; 221 (01) 130-139
  • 152 Reichman TW, Katchman H, Tanaka T. et al. Living donor versus deceased donor liver transplantation: a surgeon-matched comparison of recipient morbidity and outcomes. Transpl Int 2013; 26 (08) 780-787
  • 153 Callaghan CJ, Charman SC, Muiesan P, Powell JJ, Gimson AE, van der Meulen JHP. UK Liver Transplant Audit. Outcomes of transplantation of livers from donation after circulatory death donors in the UK: a cohort study. BMJ Open 2013; 3 (09) e003287
  • 154 Nasralla D, Coussios CC, Mergental H. et al; Consortium for Organ Preservation in Europe. A randomized trial of normothermic preservation in liver transplantation. Nature 2018; 557 (7703) 50-56
  • 155 2023 Policy Report of the U.S. Organ Procurement and Transplantation Network and the Scientific Registry of Transplant Recipients: Transplant Data 1994-2022. Department of Health and Human Services, Health Resources and Services Administration, Healthcare Systems Bureau, Division of Transplantation, Rockville, MD; United Network for Organ Sharing, Richmond, VA; University Renal Research and Education Association, Ann Arbor, MI. Available at: https://optn.transplant.hrsa.gov/media/eavh5bf3/optn_policies.pdf . Accessed July 24, 2024
  • 156 Bowlus CL, Arrivé L, Bergquist A. et al AASLD practice guidance on primary sclerosing cholangitis and cholangiocarcinoma. Hepatology 2023; 77 (02) 659-702 DOI: 10.1002/hep.32771.
  • 157 Primrose JN, Fox RP, Palmer DH. et al; BILCAP Study Group. Capecitabine compared with observation in resected biliary tract cancer (BILCAP): a randomised, controlled, multicentre, phase 3 study. Lancet Oncol 2019; 20 (05) 663-673
  • 158 Bridgewater J, Fletcher P, Palmer DH. et al; BILCAP Study Group. Long-term outcomes and exploratory analyses of the randomized phase III BILCAP study. J Clin Oncol 2022; 40 (18) 2048-2057
  • 159 Horgan AM, Amir E, Walter T, Knox JJ. Adjuvant therapy in the treatment of biliary tract cancer: a systematic review and meta-analysis. J Clin Oncol 2012; 30 (16) 1934-1940
  • 160 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 (24) 2617-2622
  • 161 Jeong H, Kim KP, Jeong JH. et al. Adjuvant gemcitabine plus cisplatin versus capecitabine in node-positive extrahepatic cholangiocarcinoma: the STAMP randomized trial. Hepatology 2023; 77 (05) 1540-1549
  • 162 Waseem D, Tushar P. Intrahepatic, perihilar and distal cholangiocarcinoma: management and outcomes. Ann Hepatol 2017; 16 (01) 133-139
  • 163 Kendall T, Verheij J, Gaudio E. et al. Anatomical, histomorphological and molecular classification of cholangiocarcinoma. Liver Int 2019; 39 (Suppl. 01) 7-18
  • 164 Rizzo A, Carloni R, Frega G. et al. Intensive follow-up program and oncological outcomes of biliary tract cancer patients after curative-intent surgery: a twenty-year experience in a single tertiary medical center. Curr Oncol 2022; 29 (07) 5084-5090
  • 165 Lamarca A, McNamara MG, Hubner R, Valle JW. Role of ctDNA to predict risk of recurrence following potentially curative resection of biliary tract and pancreatic malignancies. J Clin Oncol 2021; 39 (3, Suppl): 336
  • 166 Boerner T, Drill E, Pak LM. et al. Genetic determinants of outcome in intrahepatic cholangiocarcinoma. Hepatology 2021; 74 (03) 1429-1444
  • 167 Mirallas O, López-Valbuena D, García-Illescas D. et al. Advances in the systemic treatment of therapeutic approaches in biliary tract cancer. ESMO Open 2022; 7 (03) 100503
  • 168 Assistance Publique - Hôpitaux de Paris. Randomized Prospective Multicentric Study: Radio-Chemotherapy and Liver Transplantation Versus Liver Resection to Treat Respectable Hilar Cholangiocarcinoma. clinicaltrials.gov; 2021. Accessed January 1, 2024 at: https://clinicaltrials.gov/study/NCT02232932
  • 169 Gül-Klein S, Schmitz P, Schöning W. et al. The role of immunosuppression for recurrent cholangiocellular carcinoma after liver transplantation. Cancers (Basel) 2022; 14 (12) 2890
  • 170 Resch T, Esser H, Cardini B, Schaefer B, Zoller H, Schneeberger S. Liver transplantation for hilar cholangiocarcinoma (h-CCA): is it the right time?. Transl Gastroenterol Hepatol 2018; 3: 38
  • 171 Ahmad MU, Hanna A, Mohamed AZ. et al. A systematic review of opt-out versus opt-in consent on deceased organ donation and transplantation (2006-2016). World J Surg 2019; 43 (12) 3161-3171
  • 172 Gringeri E, Gambato M, Sapisochin G. et al. Cholangiocarcinoma as an indication for liver transplantation in the era of transplant oncology. J Clin Med 2020; 9 (05) 1353
  • 173 Jarnagin WR, Ruo L, Little SA. et al. Patterns of initial disease recurrence after resection of gallbladder carcinoma and hilar cholangiocarcinoma: implications for adjuvant therapeutic strategies. Cancer 2003; 98 (08) 1689-1700
  • 174 Soares KC, Kamel I, Cosgrove DP, Herman JM, Pawlik TM. Hilar cholangiocarcinoma: diagnosis, treatment options, and management. Hepatobiliary Surg Nutr 2014; 3 (01) 18-34
  • 175 Kim BH, Kim K, Chie EK. et al. Long-term outcome of distal cholangiocarcinoma after pancreaticoduodenectomy followed by adjuvant chemoradiotherapy: a 15-year experience in a single institution. Cancer Res Treat 2017; 49 (02) 473-483
  • 176 Tjaden C, Hinz U, Klaiber U. et al. Distal bile duct cancer: radical (R0 > 1 mm) resection achieves favorable survival. Ann Surg 2023; 277 (01) e112-e118

Zoom Image
Fig. 1 Integration of standard liver transplant care (top row) with cholangiocarcinoma-specific medical approaches (bottom row) to optimize liver transplantation process.