Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

nature reviews clinical oncology https://doi.org/10.

1038/s41571-023-00770-1

Review article Check for updates

Cholangiocarcinoma — novel
biological insights and therapeutic
strategies
Sumera I. Ilyas1,2, Silvia Affo3, Lipika Goyal4, Angela Lamarca5,6,7, Gonzalo Sapisochin8, Ju Dong Yang 9

& Gregory J. Gores 1


Abstract Sections

In the past 5 years, important advances have been made in the scientific Introduction

understanding and clinical management of cholangiocarcinoma The TIME of CCA


(CCA). The cellular immune landscape of CCA has been characterized Desmoplasia and CAFs in CCA
and tumour subsets with distinct immune microenvironments have
Molecular diagnosis using
been defined using molecular approaches. Among these subsets, the blood and bile
identification of ‘immune-desert’ tumours that are relatively devoid
Emerging molecular
of immune cells emphasizes the need to consider the tumour immune classifications of CCA
microenvironment in the development of immunotherapy approaches. Advances in the treatment
Progress has also made in identifying the complex heterogeneity and of CCA

diverse functions of cancer-associated fibroblasts in this desmoplastic Conclusions


cancer. Assays measuring circulating cell-free DNA and cell-free tumour
DNA are emerging as clinical tools for detection and monitoring of
the disease. Molecularly targeted therapy for CCA has now become a
reality, with three drugs targeting oncogenic fibroblast growth factor
receptor 2 (FGFR2) fusions and one targeting neomorphic, gain-of-
function variants of isocitrate dehydrogenase 1 (IDH1) obtaining
regulatory approval. By contrast, immunotherapy using immune-
checkpoint inhibitors has produced disappointing results in patients
with CCA, underscoring the requirement for novel immune-based
treatment strategies. Finally, liver transplantation for early stage
intrahepatic CCA under research protocols is emerging as a viable
therapeutic option in selected patients. This Review highlights and
provides in-depth information on these advances.

A full list of affiliations appears at the end of the paper. e-mail: gores.gregory@mayo.edu

Nature Reviews Clinical Oncology | Volume 20 | July 2023 | 470–486 470


Review article

Key points important in the diagnosis and monitoring of this malignancy6,7, and are
therefore also covered in this Review. Potent oncogenic drivers of iCCA
include fibroblast growth factor receptor 2 (FGFR2) gene fusions and
•• Cholangiocarcinomas (CCAs) are highly desmoplastic cancers neomorphic, gain-of-function variants of the genes encoding isocitrate
characterized by a tumour microenvironment that is poorly dehydrogenase (IDH), specifically IDH1 (refs. 8,9). Importantly, both
immunogenic, with an abundance of immunosuppressive cell types FGFR2 fusions and IDH1 mutations are targetable, and approved thera-
such as myeloid-derived suppressor cells and tumour-associated pies are now available for the targeted treatment of CCAs harbouring
macrophages. these genetic aberrations10–17. We discuss the clinical efficacy of these
molecularly targeted therapies in detail, considering that informa-
•• The desmoplasia of CCAs is associated with an abundance of tion continues to evolve regarding their efficacy and sequential use
heterogeneous cancer-associated fibroblasts (CAFs), with several within the treatment armamentarium for advanced-stage CCA. Finally,
transcriptomically diverse CAF subtypes identified. emerging information regarding the efficacy of liver transplantation
for early stage iCCAs, especially those occurring in the context of
•• Assessments of circulating tumour cells and cell-free tumour DNA chronic liver disease, are reviewed18. We hope that our focused update
are becoming important in the diagnosis and monitoring of CCA. on these particularly germane topics will be of high value to scientists
and clinicians studying CCA and caring for patients afflicted with this
•• Potent oncogenic drivers of intrahepatic CCAs (iCCAs) include complex and nefarious cancer.
fibroblast growth factor receptor 2 (FGFR2) gene fusions and
neomorphic, gain-of-function variants of isocitrate dehydrogenase 1 The TIME of CCA
(IDH1). FGFR2 fusions and IDH1 mutations are targetable, and approved Immunosuppressive barriers
molecularly targeted therapies are now available for the treatment CCAs are dense, desmoplastic tumours with a prominent stroma com-
of CCAs harbouring these genetic aberrations. prising CAFs and innate and adaptive immune cells, among other cell
types, that can have tumour-promoting or tumour-suppressive roles
•• The results of immune-checkpoint inhibitor monotherapy in patients depending upon the context19. The TIME of CCA is complex with intri-
with CCA have been disappointing, although the combination of cate and extensive crosstalk between the various stromal and immune
gemcitabine, cisplatin and durvalumab has been approved in the components19. Emerging literature suggests that the CCA microenvi-
first-line setting for the treatment of advanced-stage CCA. ronment is ‘immune cold’, with a paucity of cytotoxic immune cells and
an abundance of immunosuppressive elements, particularly myeloid
•• Liver transplantation is an emerging option for the subset of patients cells20–24. Indeed, CCAs have a decreased abundance of cytotoxic
with early stage iCCA occurring in the setting of cirrhosis. T lymphocytes (CTLs) and natural killer (NK) cells relative to adjacent
tumour-free liver tissues, whereas the density of regulatory T (Treg)
cells is increased24. Moreover, the expression of immune-checkpoint
Introduction molecules such as PD-1 and its ligand PD-L1 as well as CTLA4 is increased
Cholangiocarcinoma (CCA) is an epithelial cell malignancy of the liver on the tumour-infiltrating immune cells, indicating an immunosup-
and biliary tract. Histopathologically, this cancer is characterized by pressive milieu20,23–25. CCAs also have a high level of intratumoural
features of cholangiocyte differentiation (cholangiocytes are the epi- transcriptomic heterogeneity, probably owing to the presence of
thelial cells that line the intrahepatic and extrahepatic bile ducts). The multiple different cell types that promote immunosuppression26.
incidence of CCA is increasing in several countries globally, and given These immunosuppressive elements within the TIME (Fig. 1) facili-
its high lethality with 5-year overall survival (OS) ranging from 7% to tate tumour growth and progression, immune evasion and resistance
20%, this disease has attracted considerable scientific and clinical inter- to chemotherapy, and probably contribute to the limited efficacy of
est1,2. Indeed, several outstanding reviews focusing on CCA have been immune-checkpoint inhibitor (ICI) monotherapy in clinical trials20,27,28.
published in the past few years2,3. These reviews richly describe the Tumour-associated macrophages (TAMs) are the predominant
three anatomical subsets of the disease — namely, perihilar cholangio- immunosuppressive myeloid cell type in the TIME of CCA20,21. More­
carcinoma (pCCA), intrahepatic cholangiocarcinoma (iCCA) and distal over, an abundance of TAMs is associated with poor patient outcomes21.
cholangiocarcinoma (dCCA) — and their epidemiology, histopathology, A high ratio of monocytes to lymphocytes in the blood is an independent
genetics and molecular drivers. These recent reviews also summarize predictor of unfavourable outcomes in patients with CCA21,29–31. Further-
the clinical diagnosis, staging and treatment of CCA. Yet given a virtual more, patients with iCCAs harbouring a high percentage of macro­
explosion of published manuscripts (>6,000 publications) since the phages expressing CD206, a scavenger receptor expressed on TAMs,
last Review of CCA in this journal in 2018 (ref. 4), an update is warranted. and a low percentage expressing CD86, a co-stimulatory signal for
This update is focused on what we believe are the most perti- T cells, have a higher risk of disease recurrence and worse OS following
nent scientific and clinical advances made in the past 5 years. CCA is surgical resection than those with CD206-low, CD86-high tumours32.
a highly desmoplastic cancer characterized by a rich tumour immune TAMs mediate tumour initiation and progression via diverse path-
microenvironment (TIME)2. We highlight the deepening understand- ways, and multiple mechanisms are involved in TAM accumulation
ing of the TIME based on single-cell RNA sequencing (scRNA-seq) within the TIME. Kupffer cells, which are resident macrophages in
and immunophenotyping. The desmoplasia of CCA is the result of the liver, can facilitate cholangiocyte proliferation and oncogenic
an abundance of cancer-associated fibroblasts (CAFs)5, which have transformation by secreting TNF that triggers JNK signalling in the
not been fully described in prior reviews, and the nature and role of cholangiocellular compartment33. Accordingly, JNK inhibition attenu-
CAFs in CCA pathogenesis will be emphasized herein. Assessments ates iCCA development in preclinical models33. In addition, inflamma-
of circulating tumour cells and cell-free DNA (cfDNA) are becoming tory macrophages in the TIME promote a WNT-high state that favours

Nature Reviews Clinical Oncology | Volume 20 | July 2023 | 470–486 471


Review article

Immunosuppressive TIME Re-engineered immunogenic TIME

CCA cell
MDSC
TAM
Therapeutic strategies
• LXR agonism + anti-CSF1R antibody + ICI
TAM • CD40 agonism + ICI
• Trametinib + ICI MDSC
Treg cell • GMCSF neutralization
Treg cell
CD8+ T cell

CCA cell NK cell

CD8+ T cell CD4+ T cell

Tumour growth and progression Tumour regression

Fig. 1 | Therapeutically re-engineering the TIME of CCA to reduce tumour of granulocyte–macrophage colony-stimulating factor (GMCSF), liver
burden. The cholangiocarcinoma (CCA) tumour immune microenvironment X-receptor (LXR) agonism to inhibit MDSCs combined with macrophage
(TIME) is poorly immunogenic with an abundance of immunosuppressive inhibition using anti-colony-stimulating factor 1 receptor (CSF1R) antibodies
cell types such as myeloid-derived suppressor cells (MDSCs), tumour- and immune-checkpoint inhibitors (ICIs), as well as the combination of ICIs with
associated macrophages (TAMs) and regulatory T (Treg) cells that foster tumour CD40 agonism or MEK inhibition with trametinib. These immunotherapeutic
growth and progression. Immunotherapeutic strategies that have been strategies have the potential to re-engineer the CCA TIME to an immunogenic
investigated in preclinical studies using mouse models include neutralization one that is less favourable to the tumour. NK, natural killer.

tumour growth and progression across several preclinical models of In preclinical models of colitis or primary sclerosing cholangitis, which
CCA34. Consequently, either WNT inhibition or macrophage depletion is an important risk factor for CCA, intestinal barrier dysfunction and
markedly reduces the tumour burden in these models34. TNF-like weak gut dysbiosis enable bacterial cells and endotoxins to accumulate in
inducer of apoptosis (TWEAK) predominantly produced by immune the liver22. Consequently, hepatocytes secrete the chemokine CXCL1,
cells, including macrophages, can induce secretion of monocyte which recruits CXCR2+ PMN-MDSCs that accelerate the development
chemoattractant protein 1 (MCP1, also known as CCL2) by CCA cells of CCA22. This effect can be abrogated through antibiotic treatment
and thereby promotes macrophage recruitment to the CCA TIME and or neutralization of CXCL1 (ref. 22). These findings highlight the inter-
induces their polarization to a pro-tumour CD206+ TAM phenotype, play between the gut microbiota, intestinal barrier function and the
in association with an increase in tumour growth35. Indeed, although CCA TIME.
resident macrophages have an essential role in CCA development,
the majority of TAMs in established murine tumours are recruited Insights from multiomics analyses
from peripheral monocytes20. Expression of PD-L1 is an important Emerging single-cell and bulk multiomics studies have begun to illumi-
mechanism of tumour immune evasion, and PD-L1 expression within nate the complex heterogeneity of the CCA TIME, and its relationship
the tumour front is associated with unfavourable OS following sur- with distinct molecular subtypes of the disease and correlation with
gical resection of iCCA23. Monocyte-derived TAMs in the CCA TIME patient prognosis, as well as its modulation through immunother-
abundantly express PD-L1 and are in fact the main source of PD-L1 in apy. scRNA-seq of 144,878 cells from 14 iCCA specimens revealed two
human CCAs, and these recruited PD-L1+ TAMs are essential for CCA tumour subtypes based on differential expression of the markers S100P
progression in mouse models20. However, genetic disruption of TAM and SPP1 (also known as osteopontin)39. Each subtype had a distinct
recruitment or pharmacological inhibition of TAM function did not TIME and correlated with a different patient prognosis. For example,
reduce CCA tumour burden owing to a compensatory emergence of S100P+SPP1− iCCAs (termed perihilar large-duct-type tumours) had
another immunosuppressive myeloid cell population, myeloid-derived decreased infiltration of CD4+ T cells and CD56+ NK cells while having
suppressor cells (MDSCs). an increased abundance of CCL18+ macrophages and PD-1+CD8+ T cells
MDSCs are a heterogeneous population of immature myeloid relative to the other tumour subtype39. By contrast, S100P−SPP1+ iCCAs
cells categorized as either monocytic (M-MDSCs) or granulocytic (peripheral small-duct-type) had enhanced infiltration of SPP1+ macro­
(G-MDSCs), with the latter also referred to as polymorphonuclear phages39. Moreover, S100P−SPP1+ iCCAs were associated with a less
MDSCs (PMN-MDSCs)36. MDSCs foster tumour growth and have potent aggressive tumour phenotype (including lower levels of serum CA19-9
immunosuppressive properties that contribute to CTL and NK cell and CEA and tumour Ki67 expression, as well as a lower prevalence of
dysfunction in cancer20,36. Notably, patients with CCA have higher circu- lymph node metastasis and advanced-stage disease) and better OS
lating levels of CD14+CD11b+HLA-DR− MDSCs than individuals without (based on immunohistochemistry microarray data from 186 patients
known cancer37. Moreover, high levels of MDSCs in the peripheral blood and RNA-seq data from 66 patients)39. In another multiomics analysis
are associated with resistance to ICIs in patients with melanoma 38. of 151 iCCAs using proteomics analysis, whole-exome sequencing and

Nature Reviews Clinical Oncology | Volume 20 | July 2023 | 470–486 472


Review article

scRNA-seq, the tumours were classified into three molecular subtypes: agent alone in a mouse model of iCCA45. In aggregate, the results of
chromatin remodelling, metabolism and chronic inflammation40. The these preclinical studies support the concept that therapeutic strate-
chronic inflammation subtype was characterized by the presence of gies that modulate the CCA TIME by shifting the balance from immuno­
APOE+C1QB+ TAMs and associated with poor patient outcomes40. By suppressive factors in favour of cytotoxic elements can augment the
comparison, the metabolism subtype had frequent mutations in KMT2D antitumour response and the efficacy of ICIs (Fig. 1).
(encoding histone-lysine N-methyltransferase 2D) and was associated
with lower inflammatory activity40. Desmoplasia and CAFs in CCA
Evolving data also suggest that the CCA TIME can influence One of the main features of CCA is its highly desmoplastic nature with
response to immunotherapy. Multiplexed single-cell transcriptomic an accumulation of CAFs and excessive deposition of extracellular
and epitope sequencing of 200,000 peripheral blood mononuclear matrix (ECM) in the TIME, which contributes to the poor prognosis of
cells from nine patients with CCA and eight matched donors without patients with this disease27,46–49. CAFs intercommunicate with a wide
known cancer demonstrated high levels of a unique population of variety of cell types including cancer cells, immune cells and endothelial
CD14+ monocytes, termed CD14CTX cells, in patients with tumours resist- cells, all interacting in a multidirectional network influencing chol-
ant to anti-PD-1 antibodies41. The CD14CTX cells had increased expression angiocarcinogenesis27,50. Indeed, CAFs are one of the most abundant
of immunosuppressive cytokines and chemotactic molecules, and cell types in the TIME of CCA and contribute to tumour neoangiogen-
induced expression of SOCS3 in CD4+ T cells, thereby inhibiting their esis, immunomodulation and stiffness by secreting growth factors,
functional activity. The presence of CD14CTX cell is also of prognostic cytokines, exosomes and ECM proteins; therefore, CAFs have long
relevance, given that tumours expressing a CD14CTX gene signature been considered a putative target for therapy in CCA5,48,49,51. However,
were associated with unfavourable OS in patients with CCA as well as therapeutic targeting of CAFs in other tumour types has thus far failed
in those with other ICI-refractory cancers41. to improve patient outcomes, highlighting the need for a more metic-
ulous characterization of these cells52. Indeed, despite their overall
Overcoming the immunosuppressive TIME tumour-promoting role in CCA5,51,53, specific CAF mediators have now
Preclinical studies have provided insights into potential strategies to been shown to exert tumour-promoting or tumour-restricting effects5,
circumvent immunosuppressive barriers in the TIME of CCAs. TAMs adding more complexity to an already intricate picture.
and MDSCs seem to have an integral role in the CCA TIME; therefore,
combined targeting of both of these cell populations is a rational thera- CAF heterogeneity
peutic approach (Fig. 1). Activation of the transcription factor liver-X CAFs are heterogeneous in their origins, transcriptomes and functions
receptor (LXR) restricts the survival and abundance of MDSCs42. In a (Fig. 2), as observed in several tumour types46,48,54,55. Mainly originating
syngeneic orthotopic mouse model of CCA, pharmacological target- from resident fibroblasts in the liver, CAFs in the CCA TIME demonstrate
ing of MDSCs using either an anti-Ly6G antibody or the LXR agonist considerable plasticity; for example, becoming activated in response
GW3965 combined with targeting of TAMs using an anti-CSF1R anti- to the dynamic microenvironment during cholangiocarcinogenesis5,51.
body augmented the efficacy of anti-PD-1 antibodies, with consequent Classically identified based on expression of α-smooth muscle actin
prolongation of survival20. Neutralization of granulocyte–macrophage (α-SMA), CAFs are also characterized by expression of collagen type I
colony-stimulating factor (GMCSF) is another approach that has dem- α1 chain (COL1A1), fibroblast activation protein, platelet-derived
onstrated promise in preclinical models of iCCA21. GMCSF has an inte- growth factor receptor-α (PDGFRα) and PDGFRβ, among other pro-
gral role in myeloid cell programming, and higher expression of this teins5,26,52,54,56–59. Heterogeneity in CAF biomarkers has been confirmed
cytokine is associated with unfavourable OS in patients with iCCA fol- by data from both bulk RNA-seq and scRNA-seq transcriptomic stud-
lowing surgical resection21. Accordingly, antibody-mediated GMCSF ies, which also outline the coexistence of transcriptomically diverse
blockade curtailed tumour growth in an orthotopic model of CCA by CAF subtypes in the TIME of CCA5,57,58. Despite the lack of a common
decreasing TAM abundance and repolarizing immunosuppressive nomenclature, the presence of specific CAF subtypes enriched in genes
TAMs and MDSCs in a manner that facilitated the T cell response21. and pathways related to either a myofibroblastic or an inflammatory
CD40 is a receptor that is expressed on antigen-presenting cells, phenotype (myCAFs and iCAFs, respectively) has been confirmed in
including dendritic cells (DCs) as well as macrophages, and promotes all such studies5,57,58. Moreover, the identification of intermediate CAF
DC activation and macrophage re-education to an antitumour, inflam- populations arising from the same progenitor cell points towards the
matory phenotype43. In several different preclinical models of iCCA, existence of transient rather than static CAF subtypes, thus linking
combination of an agonistic anti-CD40 antibody with an anti-PD-1 anti- CAF heterogeneity to CAF plasticity5. Despite the abundant evidence
body markedly reduced tumour burden, in association with increases of the overall tumour-promoting role of CAFs in CCA5,51,53, specific CAF
in the abundance and activation of macrophages and DCs as well as mediators such as COL1A1 have been shown to restrain rather than
cytotoxic cell types including CD8+ T cells, NK cells and CD4+ T cells; support tumorigenesis5,56,60, thus challenging the historical paradigm
most of these cell types were functionally implicated in the antitumour and highlighting the need to further investigate the functionality of
response through depletion experiments44. Furthermore, CD40 ago- specific CAF subtypes.
nism combined with PD-1 antagonism enhanced the efficacy of gem-
citabine–cisplatin chemotherapy, with consequent increased mouse Therapeutic opportunities
survival compared to gemcitabine–cisplatin alone44. New therapeutic strategies are urgently needed to improve outcomes
Other systemic therapies can also prime the TIME and potentiate in patients with CCA, and CAFs are considered an attractive target
the response to ICIs. For example, MEK inhibition with trametinib for therapy because they actively participate in cholangiocarcino-
upregulates tumour cell expression of MHC class I (MHC I) molecules genesis3,27,46,49,59. With the emerging evidence of CAF heterogeneity,
and PD-L1. Hence, the combination of trametinib and an anti-PD-1 anti- however, targeting these cells becomes more challenging and makes
body had enhanced antitumour efficacy compared with that of either strategies aimed at their global targeting infeasible51. As evidenced

Nature Reviews Clinical Oncology | Volume 20 | July 2023 | 470–486 473


Review article

Other TME

HSC

PF

Cell of origin Crosstalk

vCAF
Tumour
restriction

CAF heterogeneity

iCAF

Subtype Function

Tumour
promotion

myCAF

apCAF

Fig. 2 | Different shades of CAF heterogeneity in CCA. Cholangiocarcinoma CAFs (vCAF), inflammatory CAFs (iCAF), myofibroblastic CAFs (myCAF)
(CCA) is a highly desmoplastic tumour type characterized by a rich stroma and antigen-presenting CAFs (apCAF). CAF heterogeneity is also reflected
with an abundance of fibroblasts and their molecular products. These cancer- in their functionality, with evidence for subtypes with tumour-promoting
associated fibroblasts (CAFs) are heterogeneous in several aspects including and/or tumour-restricting activities. Furthermore, diverse CAF subtypes are
their cell of origin, mainly arising from hepatic stellate cells (HSCs) and likely to interact differentially with tumour cells and other cells in the tumour
portal fibroblasts (PFs). Moreover, transcriptomically diverse CAF subtypes microenvironment (TME) during tumorigenesis.
have been described to coexist in the same tumour and include vascular

by data from studies reported in the past 2 years, interfering with the conditional deletion of Col1a1 in this cell type in mouse models of other
cancer cell–CAF crosstalk and the development of dual therapies desmoplastic cancers such as pancreatic ductal adenocarcinoma60,
aimed at targeting both CAFs and cancer cells seem to be the most probably by forming a physical barrier that mechanically restrains
promising therapeutic strategy5,56,57,60. Mediators of this direct cross- tumour invasion56. These opposing effects highlight the complex role
talk that promote tumour growth include hyaluronan produced by of CAFs in the TIME; a comprehensive understanding of these dual
myCAFs and hepatocyte growth factor (HGF) secreted by iCAFs, which effects would guide the development of more effective therapeutic
can stimulate cancer cells via various hyaluronan receptors and MET, strategies. For example, therapeutic strategies that target tumour-
respectively5,56. Nevertheless, myCAFs also secrete type I collagen that promoting CAF mediators (such as HGF) while maintaining the effect
can counteract these tumour-promoting effects5, as shown through of tumour-suppressive mediators (such as type I collagen) have the

Nature Reviews Clinical Oncology | Volume 20 | July 2023 | 470–486 474


Review article

potential to transform CAFs from a pro-tumour to an antitumour cell changes in CA19-9 levels (Pearson’s r = 0.69 for interval slopes, r = 0.93
population. Furthermore, stratification of CCAs based on patient- for interval differences), suggesting that ctDNA levels reflect changes in
specific mutational profiles and TIME characteristics57 might provide disease burden over time during treatment7. Furthermore, longitudinal
new insights to advance towards precision medicine. Therefore, a bet- profiling of ctDNA can be performed to track the emergence of resist-
ter understanding of CAF heterogeneity and plasticity is needed to ance to chemotherapy and, therefore, potentially facilitate the timely
develop new therapeutic approaches aimed at targeting CCA taking use of second-line or third-line treatments for CCA7,64. Thus, deter-
into consideration the complexity of its TIME. mination of cancer-derived cfDNAs present in blood is a promising
and emerging technology for the management of patients with CCAs.
Molecular diagnosis using blood and bile Confirmation of malignant biliary stricture is another major clini-
Clinical gaps exists in the early diagnosis of CCA as well as the moni- cal challenge in patients with indeterminate biliary strictures detected
toring of disease progression and treatment responses. Utilization using non-invasive imaging. The diagnostic work-up in these patients
of blood-based liquid biopsy assays has shown promise in detecting involves endoscopic retrograde cholangiopancreatography (ERCP),
multiple types of early stage cancers61,62. Among various classes of but the sensitivity of traditional methods such as ERCP-guided brush
liquid biopsy, circulating tumour DNA (ctDNA), a fraction of cfDNA cytology or biopsy sampling for the detection of malignant biliary
originating from tumour cells, has been studied most extensively stricture is <40%66,67. To address this diagnostic dilemma, molecu-
for early cancer detection. Indeed, ctDNA in the blood might serve lar characterization of biliary brush specimens and/or bile has been
as a diagnostic marker for CCA6,63. A proof-of-concept retrospective evaluated as a tool for the diagnosis of malignancy. Fluorescence in situ
study published in 2021 investigated a panel of ctDNA methyla­ hybridization (FISH) analysis of biliary brush specimens for chromo-
tion markers for the detection of CCA specifically6. The methylated some 1q21, 7p12, 8q24 and 9p21 aberrations was found to improve the
DNA markers were identified as differentially methylated regions in accuracy in diagnosing malignant biliary stricture, achieving 65%
frozen CCA specimens versus matched adjacent benign biliary epithelia sensitivity and a specificity of 93%67. Moreover, somatic KRAS mutations
and liver parenchyma tissues, and a total of nine markers were selected and/or loss-of-heterozygosity alterations across ten tumour-suppressor
from this tissue comparison6. Subsequently, this panel of methyl- genes detected in DNA isolated from biliary brush specimens have been
ated DNA markers was blindly tested in plasma cfDNA samples from shown to be excellent diagnostic biomarkers for malignant biliary
54 patients with pCCA and five with dCCA as well as from control groups strictures68. In this prospective study involving 100 patients with bil-
comprising patients with primary sclerosing cholangitis or individuals iary strictures (41% with malignancy), routine FISH identified nine of
without known liver disease (n = 22 and 95, respectively)6. The cfDNA 28 malignancies not detected on cytology analysis; mutation profil-
methylation panel had a sensitivity of 76% for the detection of CCA at ing of cell-free supernatant fluid of the brush specimen identified an
a specificity of 94%6. The panel correctly identified 64% of patients additional eight malignancies not detected by FISH, increasing the
with CCAs amenable to transplantation or surgical resection and 63% overall sensitivity to 51% versus 32% with cytology alone, whilst main-
of CCAs among patients with low serum CA19‐9 levels (≤100 U/ml)6, taining 100% specificity68. These data highlight the complementary
highlighting the potential utility of ctDNA methylation assays for the role of these diagnostic assays. Considering the rapid evolution of next-
detection of early stage CCA. In addition, methylation of OPCML and generation sequencing (NGS) capacity, a single-centre prospective
HOXD9 in cfDNA was assessed in a cohort of 40 patients and compared study in 252 patients with biliary stricture utilized BiliSeq, a targeted
with a disease control group comprising patients with non-malignant NGS panel encompassing 28 genes, to screen for mutations in biliary
biliary diseases such as cholelithiasis63. The combination of OPCML and brush cytology samples with or without biliary biopsy specimens;
HOXD9 markers had a sensitivity and specificity of 62.5% and 100%, the assay had good performance for the detection of CCA with 73%
respectively, for the diagnosis of CCA63. These findings require prospec- sensitivity and 100% specificity69. Notably, BiliSeq had a sensitivity of
tive validation in larger cohorts before methylated ctDNA markers can 83% in patients with primary sclerosing cholangitis69. For comparison,
be used for routine clinical care. the sensitivity and specificity of elevated serum CA19-9 alone were 76%
Genetic and epigenetic alterations detected in cfDNA are strongly and 69%, respectively, and for pathological evaluation alone were
correlated with those present in tumour tissue7,64; therefore, tumour- 48% and 99%69. In another prospective study, NGS-based mutational
specific somatic mutations and epigenetic changes in plasma ctDNA analysis of cfDNA from ERCP-derived bile samples using a 52-gene panel
could potentially be utilized for minimally invasive longitudinal moni- had a sensitivity of 96.4% and a specificity of 69.2% for the detection of
toring of dynamic changes in tumour biology and surveillance for CCA in 68 patients with suspicious biliary strictures70.
disease progression. In a multicentre retrospective analysis of 138 Altered DNA methylation markers in biliary brush specimens and/or
blood-derived cfDNA samples from 124 patients with CCA (70% of bile samples from patients with indeterminate biliary strictures might
whom had iCCA) using a 73-gene panel, 105 samples (76%) had one also improve the diagnosis of CCA. The first proof-of-concept case–
or more detectable genetic alterations after excluding variants of control study in patients with CCA (n = 15 and 34 in the training and
unknown significance, and 76 samples (55%) had therapeutically validation cohorts, respectively) and control patients with primary
relevant alterations, including BRAF mutations, ERBB2 (also known sclerosing cholangitis (n = 20 and 34) reported that a panel of four
as HER2) amplifications, FGFR2 fusions or mutations and IDH1 muta- methylation markers (in CDO1, CNRIP1, SEPT9 and VIM) measured
tions65. Thus, ctDNA could be a valuable resource for tumour molecular in DNA from biliary brush specimens had a sensitivity of 85% and a
profiling, especially when obtaining tumour tissue samples is techni- specificity of 98% for the detection of CCA (area under the receiver
cally challenging or unsafe, which is a major challenge in managing operating characteristic curve (AUC) 0.944)71. Several smaller
CCA. In addition, ctDNA can be used to monitor response to chemo- case–control studies have shown the outstanding performance of
therapy and targeted therapy. A proof-of-concept study of serial plasma DNA methylation biomarkers (for example, at loci associated with
cfDNA testing in eight patients with pancreaticobiliary cancers demon- EMX1 or HOXD8) in biliary brush and/or bile samples, with AUC val-
strated that changes in cfDNA mutant allele fraction correlate well with ues of 0.98–1 for the detection of malignant biliary strictures6,72.

Nature Reviews Clinical Oncology | Volume 20 | July 2023 | 470–486 475


Review article

Similarly, profiling of microRNAs in bile samples has been investigated, class was characterized by activation of oncogenic signalling pathways,
with promising results for the detection of CCA (AUC 0.652, 0.730, KRAS/BRAF mutations, and gene expression signatures previously asso-
0.765, 0.975 and 0.975 for miR-92a-3p, miR-30d-5p, miR-944, miR-9 ciated with poor outcomes in patients with hepatocellular carcinoma78.
and miR-145*, respectively)73,74. Patients with proliferation class iCCAs had significantly worse OS than
The data from these initial biomarker studies have revealed the patients with inflammation class tumours (median 24.3 months versus
great promise of ctDNA as a novel biomarker for CCA detection. We 47.2 months; P = 0.048)78. Similarly, an integrated genomic analysis of
anticipate that such liquid biopsy assays will further improve the diag- pCCA and dCCA tumour specimens (n = 189) enabled the definition
nosis of CCA in routine clinical practice once larger-scale external of four distinct classes: metabolic, proliferation, mesenchymal and
validation studies are completed in the near future. Given that compre- immune77. Tumours of the metabolic class (comprising 19% of cases)
hensive molecular profiling is recommended in the routine clinical care had a hepatocyte-like phenotype with enrichment of genes related to
of patients with advanced-stage CCA, ctDNA analysis might also serve bile acid metabolism77. dCCAs were more likely to be categorized into
as a minimally invasive test for the selection of molecularly targeted the proliferation class (accounting for 23% of tumours overall), which
treatments. Plasma ctDNA will be particularly helpful in identifying was defined by mTOR signalling, HER2 alterations and enrichment for
patients with CCAs harbouring IDH1 mutations or FGFR2 fusions, given MYC targets77. Patients classified in the mesenchymal class (47%) had
the availability of approved therapies predicated on these alterations65. unfavourable OS and enrichment of gene signatures associated with
In an NGS-based analysis involving 1,671 patients with advanced-stage epithelial-to-mesenchymal transition77. The immune class (11%) was
CCA, genetic alterations in cfDNA were detected in samples from 84%, characterized by high levels of lymphocyte infiltration and PD-1/PD-L1
with targetable alterations in cfDNA detected in samples from 44%75. expression, as well as other transcriptomic features associated with
The concordance between cfDNA and tumour tissue analysis for the increased responsiveness to ICI in other tumour types77.
detection of IDH1 and BRAFV600E mutations was 87% and 100%, respec- The increasing emphasis in oncology on immunotherapeutic
tively, but was only 18% for FGFR2 fusions75. Although molecular analy- approaches has highlighted the integral role of the TIME and the
ses of blood, biliary brush and/or bile specimens can facilitate the potential clinical significance of immune-based classifications of CCA.
implementation of precision medicine in patients with CCA, important A TIME-based classification developed using transcriptomics from a
challenges continue to limit widespread clinical use of such assays. training set comprising 198 iCCA specimens identified four immune
First, numerous methods can be used to detect methylation markers subclasses associated with distinct immune escape mechanisms and
or mutations in cfDNA, with substantial variation in sample type (for patient outcomes: immune-desert, immunogenomic, myeloid and mes-
example, biliary brush cytology versus cell-free supernatant fluid enchymal79. This classification was validated using an independent set
versus bile), handling and processing, and analysis. Standardized of 368 iCCAs and through immunohistochemical analysis of a further
ctDNA assay protocols that can capture the full spectrum of muta- 64 iCCA samples79. In alignment with the concept that the CCA TIME is
tion and methylation with high sensitivity and specificity need to be ‘immune cold’, the dominant subclass (present in approximately 45% of
developed and validated in order to ensure robust and generalizable patients overall) was the immune-desert subset characterized by low
results. Second, most current supporting evidence comes from small- expression of TIME gene signatures79 (Table 1). By contrast, the immuno­
cohort, single-centre, case–control studies. Given the low incidence of genomic subclass was enriched for infiltration of adaptive and innate
CCA, these issues necessitate large-scale, collaborative, multicentre immune cells and activation of inflammatory and immune-checkpoint
prospective studies using standardized protocols. pathways, and was associated with the most favourable patient out-
comes79. The myeloid subset featured enrichment of monocyte-derived
Emerging molecular classifications of CCA and myeloid gene signatures, whereas the mesenchymal subclass
The prevalence of molecular profiling has enabled the definition of was enrichment for fibroblast gene signatures79. Similarly, a Stroma,
transcriptome-based classes of iCCA and pCCA76–78. Importantly, the Tumour and Immune Microenvironment (STIM) classification of iCCA,
molecular subclasses might predict patient outcomes or response to which as its name implies integrates stromal, tumour and immune
certain therapeutic approaches. For example, a genomic and tran- microenvironmental elements, categorized the preponderance of
scriptomic characterization of 104 surgically resected iCCA speci- patients into non-inflamed tumour classes (65%)57. Of the five STIM
mens revealed two distinct tumour classes associated with different classes of iCCA identified (Table 1), two inflamed classes were defined as
clinical outcomes and with the absence or presence of mutations in ‘immune classical’ (accounting for 10% of patients) and ‘inflammatory
KRAS or BRAF76. Specifically, the 5-year OS in 51 patients with ‘cluster 1’ stroma’ (~25%), with the latter characterized by a rich stroma (extensive
class tumours according to a transcriptomic classifier based on 238 desmoplasia), T cell exhaustion and KRAS mutations57. The three non-
significantly differentially expressed genes was 72% compared with inflamed classes are: (1) the ‘desert-like’ class (20%) with a paucity of
30% in 53 patients with cluster 2 tumours (P < 0.0007)76. Patients immune cells but enrichment of Treg cells; (2) the ‘hepatic stem-like’
with cluster 2 tumours also had a shorter median time to recurrence class (35%) with abundant M2-like macrophages and enrichment for
(13.7 ± 6.3 months versus 22.7 ± 18.1 months; P = 0.001)76. Integration IDH and BAP1 mutations and FGFR2 fusions; and (3) the ‘tumour clas-
of KRAS/BRAF mutational status with the 238-gene classifier grouped sical’ class (10%) characterized by cell cycle pathway activation and
all 18 evaluable patients with such mutations into the poor prognosis associated with unfavourable patient outcomes57. Although these
cluster 2 (ref. 76). Integrative molecular analyses have also been applied classifications assist in the broad categorization of CCA subtypes and
for the detection of biological classes of CCA with differential activa- support the notion that CCAs typically have an immunosuppressive
tion of certain signalling pathways. One such analysis of 149 iCCAs TIME, their clinical utility is currently unclear. Prospective validation
defined two principal tumour classes: an inflammation class (38%) of these classifications is needed before they can be used in routine
and a proliferation class (62%)78. Features of the inflammation class patient care. For example, clinical trials stratifying patients based on
included activation of inflammatory signalling pathways, overexpres- these classifications with correlation to particular treatments would
sion of cytokines and activation of STAT3, whereas the proliferation help to clarify their potential utility in the management of CCA.

Nature Reviews Clinical Oncology | Volume 20 | July 2023 | 470–486 476


Review article

Table 1 | Immune classifications of iCCA based on RNA-sequencing data

Subclass Proportion of iCCAs (%) Key features Prognostic association


(median OS, months)

Classification based on immune gene expression signatures79


Immune-desert 46–48 TME signatures with weak expression of immune and myofibroblast 42
signatures
Immunogenomic 9–13 Signatures of recruited innate immune cells, adaptive immune cells and 73
activated fibroblasts
Activation of inflammatory pathways
Myeloid 13–19 Strong expression of monocyte-derived and/or other myeloid signatures 25
Low expression of lymphoid signatures
Mesenchymal 22–28 Strong expression of fibroblast signatures 19
Stroma, Tumour and Immune Microenvironment (STIM) classification 57

Immune classical ~10 High immune infiltration (63%) –a


Moderate stromal infiltration (27%)
Inflammatory stroma ~25 Moderate immune infiltration (43%) –
High stromal infiltration (50%)
Abundant desmoplastic reaction and ECM deposition
High stiffness
Activated inflammatory stroma
T cell exhaustion
Hepatic stem-like ~35 Low immune infiltration (28%) –
Low stromal infiltration (17%)
Abundant tumour-promoting macrophages
Tumour classical ~10 Low immune infiltration (22%) –
Low stromal infiltration (12%)
Activation of cell cycle pathways
Desert-like ~20 Low immune infiltration (22%) –
Low stromal infiltration (11%)
Regulatory T cell enrichment
ECM, extracellular matrix; iCCA, intrahepatic cholangiocarcinoma; OS, overall survival; TME, tumour microenvironment. aIn a multivariate analysis, the STIM classes were not independent
predictors of outcome.

Advances in the treatment of CCA The identification of actionable genetic alterations in approxi-
Given that the objective of this Review is to provide an overview of perti- mately 40–50% of iCCAs and 15–20% of pCCAs and dCCAs88 has trans-
nent clinical updates since our last Review in the journal4, here we focus formed the therapeutic landscape for patients with advanced-stage CCA.
on progress made over the past 5 years in systemic and non-systemic Mutational profiling is usually performed on tumour tissue specimens,
therapeutic strategies for CCA. Locoregional therapies including radio- but given failure rates of up to 27% in CCA biopsies, liquid biopsy
therapy, transarterial chemoembolization and radioembolization analysis of ctDNA might serve as an additional tool for identifying
were highlighted in our prior Review4. Since then, no major advances therapeutic targets, as described above64,65,89. Genetic alterations
in locoregional treatments for CCA have been made, and thus such that define the eligibility of patients with CCA for molecularly tar-
therapies are not included in this update. geted therapies based on FDA approvals or recommendations in the
National Comprehensive Cancer Network (NCCN) guidelines are
Molecularly targeted therapies discussed below90.
Several high-throughput genomic sequencing analyses have eluci-
dated the distinct mutational landscapes of iCCA, pCCA and dCCA, thus FGFR2 fusions and other rearrangements. FGFR signalling has key
revealing differences in biology between these anatomical subtypes roles in various physiological processes and drives cell proliferation,
of the disease80–84. Actionable alterations such as FGFR2 fusions and growth and survival, thus making this pathway susceptible to hijack by
IDH1 mutations occur almost exclusively in iCCAs, whereas pCCAs cancer cells91,92. FGFR2 fusions and other rearrangements, which were
and dCCAs more commonly harbour HER2 amplifications and KRAS first reported in CCAs in 2013 and subsequently found to be present
mutations80–84. Mutation frequencies among CCAs vary geographi- in 13–14% of iCCAs93–95, constitute one of the most highly actionable
cally and by aetiology, with Asian populations having lower fre- alterations in this disease. The first two molecularly targeted therapies
quencies of IDH1 mutations than non-Asian populations, and liver to gain regulatory approval for the treatment of CCA were the ATP-
fluke-associated CCAs having a lower prevalence of IDH1 mutations competitive, reversible FGFR inhibitors pemigatinib and infigratinib13,14.
and a higher prevalence of TP53 mutations than those with other These oral small-molecule inhibitors demonstrated overall response
aetiologies85–87. rates (ORRs) of 35.5% and 23.1%, respectively, in single-arm phase II

Nature Reviews Clinical Oncology | Volume 20 | July 2023 | 470–486 477


Review article

trials involving patients with previously treated advanced-stage CCAs FGFR2 inhibitor that has activity against primary FGFR2 alterations
harbouring FGFR2 fusions or rearrangements10,11 (Table 2), resulting and common resistance mutations106. In a first-in-human study involv-
in FDA Accelerated Approval for this indication in April 2020 and May ing 35 patients with FGFR2-altered CCA, this agent had encouraging
2021, respectively13,14. However, the development of infigratinib has clinical activity, with >10% tumour shrinkage observed in nine (56%)
been halted according to a manufacturer’s press release96. A third selec- of 16 patients with CCA previous treated with an FGFR inhibitor106.
tive FGFR inhibitor, futibatinib, binds covalently and irreversibly to the Other FGFR inhibitors under investigation in patients with CCA
P-loop cysteine residue in the FGFR kinase domain and achieved an ORR include the pan-FGFR inhibitors erdafitinib (NCT02699606) and KIN-3248
of 41.7%, with responses lasting a median of 9.7 months, in a similar (which has activity against FGFR2 gatekeeper, molecular brake and activa-
phase II study involving patients with previously treated advanced-stage tion loop mutations105; NCT05242822), the multikinase inhibitor derazan-
FGFR2-rearranged iCCA12. These data led to FDA Accelerated Approval of tinib (NCT03230318), the bivalent FGFR1–3 inhibitor tasurgratinib (E7090,
futibatinib for this indication in September 2022 (ref. 15). The safety and which blocks FGF–FGFR interactions; NCT04238715), and the FGFR1–3
efficacy of derazantinib, an FGFR1–3 inhibitor, has also been assessed inhibitor HMPL-453 (NCT04353375). Moreover, phase III trials comparing
in a phase II trial involving 103 patients with advanced-stage CCA har- the approved FGFR inhibitors against frontline gemcitabine and cisplatin
bouring an FGFR2 fusion who had received at least one line of prior chemotherapy are currently ongoing (pemigatinib (NCT03656536),
treatment97. In this study, derazantinib was associated with an ORR of infigratinib (NCT03773302) and futibatinib (NCT04093362)), as are early
21.4%, a disease control rate of 74.8% and a median OS of 15.5 months97. phase trials of combination therapies including FGFR inhibitors (gemcit-
Acquired resistance to FGFR inhibitors limits the effectiveness abine and cisplatin plus pemigatinib or ivosidenib (NCT04088188) and
of these agents, which are consistently associated with median pro- derazantinib plus atezolizumab (NCT05174650)).
gression-free survival (PFS) durations of 7–9 months10–12,97 (Table 2).
Polyclonal secondary mutations in the FGFR2 kinase domain are a IDH1 mutations. R132-mutant IDH1 acquires a neomorphic activity
common resistance mechanism98–101. Futibatinib has shown potent that converts α-ketoglutarate to 2-hydroxyglutarate, an oncome-
preclinical activity against many of these mutations and has shown tabolite that inhibits histone and DNA demethylases and results in
clinical activity after progression of CCA on treatment with reversible widespread epigenetic alterations and oncogenesis107. The frequency
FGFR inhibitors101–103. Additional next-generation inhibitors, such as the of IDH1 mutations in CCAs varies widely across studies and was found to
FGFR2-selective inhibitor RLY-4008 (NCT04526106) and the multiki- be 13.1% in iCCAs and 0.8% in pCCAs and dCCAs in a systematic review of
nase inhibitor tinengotinib (also known as TT-00420; NCT04919642), 45 studies85. Ivosidenib, an orally administered small-molecule inhibi-
are currently under investigation to assess their efficacy in overcoming tor of mutant IDH1, gained FDA approval for the treatment of chem-
resistance to prior FGFR inhibitors104–106. RLY-4008 is a highly selective orefractory IDH1-mutated CCA in August 2021 based on the results of

Table 2 | Efficacy of molecularly targeted therapies in patients with CCA

Agent Study Number of ORR (%) DCR (%) Median PFS (months) Median OS (months) Ref.
phase patientsa

FGFR inhibitors
Pemigatinibb II 107 35.5 82.2 6.9 NA 10
Infigratinibb
II 108 23.1 84.3 7.3 12.2 11
Futibatinibb II 103 41.7 82.5 9.0 21.7 12
Derazantinib I/II 103 21.4 74.8 8.0 15.5 97
IDH inhibitors
Ivosidenibb III 124 2.4 53.2 2.7 10.8 16
BRAF inhibitors
Dabrafenib plus trametinib (MEK inhibitor)b,c II 43 46.5 85.4 9.0 14.0 111
Vemurafenib c
II 9 33.3 NA NA NA 113
HER2-targeted agents
Trastuzumab plus pertuzumab II 39 23.1 51.3 4.0 10.9 115
(monoclonal antibodies)c,d
Trastuzumab deruxtecan (antibody–drug II 24 36.4 81.8 4.4 7.1 116
conjugate)c
Neratinib (pan-ErbB tyrosine kinase inhibitor)c II 25 (11 with 16 28 2.8 (1.4 in CCA subgroup) 5.4 (5.4 in CCA subgroup) 118
CCA)
Zanidatamab (bispecific antibody)c I 17 47 65 NA NA 119
CCA, cholangiocarcinoma; DCR, disease control rate; NA, not available; ORR, objective response rate; PFS, progression-free survival. aNumber of patients with target molecular aberrations:
FGFR2 fusions and/or rearrangements for FGFR inhibitors, IDH1 R132 mutations for IDH inhibitors; BRAFV600E mutations for BRAF inhibitors; and ERBB2 (also known as HER2) amplification and/or
overexpression for HER2-targeted therapies. bFDA approved for use in patients with previously treated advanced-stage CCAs harbouring the target oncogenic alteration. cData for these agents
come from patients with CCAs as well as other biliary tract cancers. dListed in National Comprehensive Cancer Network guidelines as subsequent-line therapy for patients with advanced-stage
CCA following disease progression on recommended initial treatements90.

Nature Reviews Clinical Oncology | Volume 20 | July 2023 | 470–486 478


Review article

a phase III trial showing an improvement in PFS over placebo (median in 17 patients with advanced-stage HER2-positive BTCs119 (Table 2),
2.7 versus 1.4 months; HR 0.37, 95% CI 0.25–0.54; P < 0.0001)16,17 and this agent is being further evaluated in studies including patients
(Table 2). OS was not significantly improved (HR 0.79, 95% CI 0.56–1.12; with CCAs and other BTCs (NCT04466891 and NCT03929666). Other
P = 0.093); however, the fact that 70% of patients in the placebo therapeutic strategies currently being evaluated in trials involving
group crossed over to receive ivosidenib following disease progres- patients with HER2-overexpressing CCAs include the combination of
sion is likely to have confounded this result16. Additional IDH1 inhib­ trastuzumab and the HER2 TKI tucatinib (NCT04579380) and novel
itors under investigation in patients with CCA include olutasidenib antibody–drug conjugates (A166 (NCT03602079) and zanidatamab
(FT-2102; NCT03684811), LY3410738 (NCT04521686) and HMPL-306 zovodotin (NCT03821233)).
(NCT04762602). Given that IDH1 mutations can lead to deficiencies in
the repair of double-stranded DNA damage108,109, several ongoing trials NTRK fusions. NTRK fusions occur in approximately 0.2% of CCAs
of poly(ADP-ribose) polymerase (PARP) inhibitors are also including and are highly actionable alterations in solid tumours120. These NTRK
patients with IDH1-mutant CCA (olaparib (NCT03212274). olaparib fusions are therapeutically actionable using the TRK inhibitors laro-
plus the ATR inhibitor ceralasertib (NCT03878095) and olaparib plus trectinib and entrectinib, which received FDA approval agnostic of
durvalumab (NCT03991832)). tumour histology based on efficacy across cancer types (summary ORR
of 75% with larotrectinib and 57% with entrectinib); partial responses
BRAFV600E mutations. BRAFV600E mutations occur in up to 5% of bili­ were observed in two of three patients with CCA included in the pivotal
ary tract cancers (BTCs), primarily in iCCAs110. Treatment with the trials of these agents121–124.
BRAF inhibitor dabrafenib in combination with the MEK inhibitor
trametinib led to an ORR of 47% and a median PFS of 9.0 months in RET fusions. RET fusions are also very rare in CCAs, with a prevalence
the BTC cohort of the phase II ROAR basket trial111 (Table 2). In June of 0.15% in iCCA and 0.11% in pCCA/dCCA125. The phase I/II ARROW
2022, this combination gained FDA approval for use in patients with trial evaluated the potent, selective of RET inhibitor pralsetinib in
previously treated non-colorectal solid tumours, thus including CCAs, 29 patients with advanced-stage RET-altered solid tumours126. Of the
harbouring a BRAFV600E mutation and no satisfactory alternative treat- three patients with CCA included in this basket study cohort, two had a
ment options, based on data from ROAR and several other trials112. Data partial response and one had stable disease126, gaining pralsetinib a list-
on BRAF inhibitors as monotherapy for BTCs are limited, although ing on the NCCN guidelines90. The safety and efficacy of selpercatinib,
vemurafenib induced partial responses in three of nine patients with another highly selective RET inhibitor, are being evaluated in patients
BRAFV600E-mutant BTCs in another basket trial113. with RET fusion-positive cancers, including two patients with CCA,
in the ongoing phase I/II LIBRETTO-001 basket trial127.
HER2 alterations. HER2 belongs to the ErbB family of receptor
tyrosine kinases and is overexpressed more frequently in pCCAs and Immunotherapies
dCCAs (17.4%) and gallbladder cancers (19.1%) than in iCCAs (4.8%)114. The role of ICI-based immunotherapy for the treatment of CCA has
The combination of two anti-HER2 antibodies, trastuzumab and changed drastically over the past few years128. It is important to differ-
pertuzumab, achieved an ORR of 23% and a median PFS of 4.0 months entiate the role of immunotherapy for CCA according to two separate
in 39 patients with HER2-amplified and/or overexpressing BTCs in the scenarios129.
phase IIa MyPathway basket study115 (Table 2), gaining a listing in The first scenario is related to exploration of immunotherapies
the NCCN guidelines as a recommended therapy for previously treated in a tumour-agnostic manner, whereby selection of patients relies on
HER2-positive BTCs90. The antibody–drug conjugate trastuzumab specific molecular alterations, regardless of the primary tumour site
deruxtecan demonstrated an ORR of 36.4% (including two complete or histology. In this regard, the ICI pembrolizumab is indicated for use
responses (CRs)), a median PFS of 4.4 months and a median OS of in patients harbouring CCAs with deficient mismatch repair (dMMR)
7.1 months in a similar phase II trial population (n = 22)116, and a partial and/or high microsatellite instability (MSI-H)130–132. Pembrolizumab,
response was also seen in one of eight patients with HER2-low disease. an anti-PD-1 antibody, had an ORR of 53% (21% CR rate) in a cohort of
Trastuzumab deruxtecan is being further explored in patients with 86 patients with advanced-stage dMMR solid tumours; four patients
BTCs in an ongoing phase II trial (NCT04482309). Another phase II with CCA were included in this cohort and one had a CR, with the
trial evaluated the efficacy of combination folinic acid, 5-fluorouracil other three having stable disease130. An updated analysis of the multi­
and oxaliplatin (FOLFOX) chemotherapy plus trastuzumab in cohort phase II KEYNOTE-158 study of pembrolizumab in patients with
the second-line setting in 34 patients with HER2-positive BTCs; the advanced-stage MSI-H/dMMR non-colorectal cancers included data
ORR was 29.4%, median PFS was 5.1 months and median OS was from 22 patients with CCA131. In the CCA cohort, pembrolizumab was
10.7 months117. associated with an ORR of 40.8% (CR rate 13.5%), a median response
Additionally, the oral, irreversible, pan-ErbB tyrosine kinase duration of 30.6 months, a median PFS of 4.2 months and median OS of
inhibitor (TKI) neratinib has been evaluated in patients with previ- 19.4 months, with a 3-year OS of 30.3%131. In addition, ICIs have notable
ously treated HER2-mutant CCA (as opposed to merely HER2-posi- activity against cancers with a high tumour mutational burden (TMB).
tive disease) as part of the SUMMIT basket trial118. Results from the Indeed, pembrolizumab has also been granted histology-agnostic
phase II BTC cohort of this trial, which included 11 patients with CCAs, approval by the FDA for patients with a high TMB (≥10 mutations
demonstrated an ORR of 16% in all 25 patients evaluated, with a median per megabase)133, based on a reported ORR of 29% (CR rate 4%) in a
PFS of 2.8 months and median OS of 5.4 months (1.4 months and cohort of 102 patients with previously treated advanced-stage solid
5.4 months, respectively, in the CCA subgroup)118 (Table 2). tumours (not specified how many had CCA); responses were dura-
The novel bispecific anti-HER2 antibody zanidatamab, which ble, lasting ≥12 months in 57% of patients and ≥24 months in 50%134.
targets the same epitopes as trastuzumab and pertuzumab, achieved Unfortunately, these tumour-agnostic approaches are applicable to
an interim ORR of 47% and a median response duration of 6.6 months only a very limited proportion of patients with CCA, given that <5% of

Nature Reviews Clinical Oncology | Volume 20 | July 2023 | 470–486 479


Review article

this patient population have dMMR/MSI-H or TMB-high tumours129. Interestingly, subgroup analyses did not reveal significant differences
Therefore, developing and applying immunotherapies in a way that in outcomes depending on PD-L1 expression143, primary tumour site145,
would enable a wider spectrum of patients to benefit is of substantial disease status (that is, initially unresectable versus recurrent)146 or
importance. geographical area (Asian versus rest of the world)147. On the basis of
Indeed, the second scenario is the development of immuno- these results, the FDA approved the combination of gemcitabine, cispl-
therapy in unselected populations of patients with advanced-stage atin and durvalumab in 2022 (ref. 148), and the latest NCCN guidelines
CCA. This approach is mainly based on translational research work for hepatobiliary cancer (version 1.2022)90 recommend this regimen
suggesting a potential role for immune-directed therapies in the as the preferred first-line treatment option for advanced-stage CCA.
treatment of CCA and other BTCs, considering that markers of a pre- Similarly, the KEYNOTE-966 trial, the largest phase III trial in patients
existing antitumour T cell response are generally associated with a with CCA to date, evaluated the combination of chemotherapy and
favourable prognosis in patients with these cancers135–138. Hence, first ICI therapy in the first-line treatment of patients with unresectable,
efforts in this regard were mainly focused on the use of monotherapy locally advanced or metastatic CCA or gallbladder cancer149. In this
approaches with ICIs. Unfortunately, this strategy had limited efficacy randomized, double-blind trial, 533 patients were allocated to receive
in patients with BTCs, with an ORR of 5.8%, a median PFS of 2.0 months pembrolizumab plus gemcitabine and cisplatin and 536 patients to
and median OS of 7.4 months with single-agent pembrolizumab in the receive placebo plus gemcitabine and cisplatin149. The primary end
phase II KEYNOTE-158 study28. Moreover, the efficacy was not clearly point was median OS, which was 12.7 months in the pembrolizumab
better in patients with PD-L1-positive tumours (ORR 6.6% and 13.0% plus gemcitabine and cisplatin group compared with 10.9 months in the
in KEYNOTE-158 and KEYNOTE-028, respectively)28. Given these find- placebo group (HR 0.83, 95% CI 0.72–0.95; one-sided P = 0.0034)149. In
ings, alternative approaches have been pursued, mainly in the form of contrast to the TOPAZ-1 trial, maintenance gemcitabine was allowed
combining ICIs with cytotoxic chemotherapy. in both arms without a maximum duration while cisplatin was admin-
Preliminary results from a retrospective study by Sun and col- istered for a maximum of eight cycles149. This may have accounted for
leagues139 suggested better outcomes with the addition of anti-PD-1 the ORR being the same in both arms (29%)149. On the basis of these two
antibodies to chemotherapy (38 patients) than with anti-PD-1 anti- trials, the role of ICIs in the first-line treatment of advanced-stage CCA
bodies alone (20 patients) or chemotherapy alone (19 patients); the has been firmly established.
median PFS durations were 5.1 months, 2.2 months and 2.4 months, Several other clinical trials of ICIs are ongoing in patients with
respectively, and the median OS was 14.9 months versus 4.1 months BTCs128. Beyond their addition to chemotherapy, combinations of
versus 6.0 months. Additional phase II studies in patients with previ- ICIs with TKIs have also been tested, albeit early results have been
ously untreated advanced-stage BTCs supported this rationale140–142. In mediocre. For example, a small-cohort phase II study found an ORR
the largest of these phase II studies, Oh and colleagues142 recruited 124 of only 10% with the combination of lenvatinib plus pembrolizumab in
patients to three different treatment arms: (1) chemotherapy (gemcit- patients with previously treated advanced-stage BTCs150. Nevertheless,
abine plus cisplatin) followed by concurrent chemotherapy and dual ICI other ongoing clinical trials are exploring this potential alternative,
therapy with the anti-PD-L1 antibody durvalumab and the anti-CTLA4 chemotherapy-free ICI-based therapeutic strategy (NCT04211168).
antibody tremelimumab from cycle 2 (30 patients); (2) concurrent Thus, the future of immunotherapy for CCA looks bright for the
chemotherapy plus durvalumab from cycle 1 (47 patients); or (3) con- first time. Efforts to identify predictive biomarkers for patient selection
current chemotherapy plus durvalumab and tremelimumab from and to understand the mechanisms of resistance will be paramount in
cycle 1 (47 patients). Whenever tremelimumab was administered, a order to realize the full potential of ICIs and other immunotherapies.
maximum of four cycles were given. The primary end point of the study
was ORR, which was highest in the arms in which ICIs were administered Liver transplantation for iCCA
from cycle 1 onwards (ORRs of 50%, 72% and 70% in arms 1, 2 and 3, Transplant oncology is a developing field that merges the discip­lines
respectively); however, the median PFS was similar between arms of organ transplantation and oncology151. iCCA is one of the para-
(12.8 months, 11.8 months and 12.3 months, respectively)142. In view digms of transplant oncology, but remains a formal contraindication
of a more favourable toxicity profile without compromised efficacy for liver transplantation outside clinical trials in most jurisdictions.
in the arm without tremelimumab, the combination of gemcitabine, Two distinct populations of patients with iCCA might benefit from
cisplatin and durvalumab was selected for further assessment in the liver transplantation: (1) patients with decompensated cirrhosis who
phase III TOPAZ-1 trial. have a liver mass with imaging characteristics that are not diagnostic
In TOPAZ-1, 685 patients with treatment-naive advanced-stage of hepatocellular carcinoma and are found to have iCCA following
CCAs or gallbladder cancer were randomly assigned to receive gem- biopsy sampling; and (2) those with multifocal and large iCCAs usually
citabine and cisplatin plus either durvalumab or placebo143. The occurring in otherwise healthy livers.
primary end point was OS, which was significantly improved with In the past 10 years, enthusiasm to incorporate iCCA as an indi-
durvalumab (median 12.8 months versus 11.5 months in the placebo cation for liver transplantation has increased after data from sev-
group; 24-month OS 24.9% versus 10.4%; HR 0.80, 95% CI 0.66–0.97; eral retrospective studies demonstrated excellent outcomes in
P = 0.021)143. The durvalumab group also had improvements in PFS patients with cirrhosis and with ‘very early’ and ‘early’ iCCAs (defined
(median 7.2 months versus 5.7 months; HR 0.75, 95% CI 0.63–0.89; as single tumours ≤2 cm and ≤5 cm, respectively) who received a liver
P = 0.001) and ORR (26.7% versus 18.7%; OR 1.60, 95% CI 1.11–2.31)143. transplant (Table 3). Most of these studies involved patients either
This improved activity was reported without additional toxicity (for initially misdiagnosed with hepatocellular carcinoma or with an inci-
example, grade 3−4 adverse events in 75.7% versus 77.8% of patients)143, dental iCCA mass found on explant pathology. A prospective phase II
and with a trend towards a longer time to deterioration in patient- trial of liver transplantation in patients with cirrhosis and very early
reported global health status according to EORTC QLQ C30 scores iCCA (NCT02878473) is now underway (Table 3); however, accruing
favouring the durvalumab arm (HR 0.87, 95% CI 0.69–1.12; P = 0.279)144. patients has been challenging owing to the difficulty of listing such

Nature Reviews Clinical Oncology | Volume 20 | July 2023 | 470–486 480


Review article

Table 3 | Summary of key contemporary studies of liver transplantation in patients with iCCA

Study (year of Tumour size iCCAs discovered Patients OS RFS Recurrence rate Chemotherapy
publication) (n) incidentally/initially with treatment
misdiagnosed as HCC cirrhosis (n)
Retrospective studies in patients with incidental or misdiagnosed small iCCAs, mainly associated with cirrhosis
Sapisochin Single ≤2 cm 23 (79%)/6 (21%) 29 (100%) Overall: 79% at 1 year; NR Overall: 11% None
et al. (2014)156 (8) 61% at 3 years; 45% at at 1 year; 29% at
Multiple or 5 years. 3 years; 29% at
single >2 cm ≤2 cm tumour group: 5 years.
(21) 100% at 1 year; 73% at ≤2 cm tumour
3 years; 73% at 5 years. group: 0% at 1 year;
>2 cm tumour group: 71% 0% at 3 years;
at 1 year; 43% at 3 years; 0% at 5 years.
34% at 5 years >2 cm tumour
group: 26% at
1 year; 42% at
3 years; 42%
at 5 years
Sapisochin Single >2 cm 8 (24%)/25 (76%) 33 (100%) 1 year: 79% NR 1 year: 30% None
et al. (2016)157 or multiple 3 years: 50% 3 years: 47%
tumours of any
5 years: 45% 5 years: 61%
size (33)
Single ≤2 cm 7 (47%)/8 (53%) 15 (100%) 1 year: 93% NR 1 year: 7% None
(15) 3 years: 84% 3 years: 18%
5 years: 65% 5 years: 18%
Jung et al. Single ≤2 cm 1 (6%)/14 (88%) 12 (13%) 1 year: 81% (63%a) NR 1 year:56% (50%a) None
(2017)158 (4) 3 years: 52% (63%a) 3 years:56% (50%a)
Multiple ≤2 cm 5 years: 52% 5 years:78%
(2)
Single >2 cm
(10)
De Martin et al. Largest nodule 5 (10%)/44 (90%) 49 (100%) 1 year: 90% (88%b, 92%c) 1 year: 87% NR None
(2020)159 ≤5 cm (49; 24 3 years: 76% (65%b, 87%c) (81%b, 78%c)
patients with 3 years: 79%
5 years: 67% (65%b,
iCCA only (74%b, 69%c)
69%c)
combined with
5 years: 75%
25 patients
(74%b, 55%c)
with mixed
HCC–iCCA
for analyses)
Retrospective studies in patients with large multifocal iCCAs diagnosed prior to liver transplantation
Lunsford et al. Among 6 NA 1 (17%) 1 year: 100% 1 year: 50% 1 year: 50% Yes, neoadjuvant
(2018)160 patients: 3 years: 83.3% 3 years: 50% 3 years: 50% and adjuvant
median
5 years: 83.3% 5 years: 50% 5 years: 50%
number of
tumours 4
(IQR 3.0–5.8);
median
cumulative
diameter
10.5 cm (IQR
7.0–13.5 cm)

Ito et al. 22 of 31 NA 14 (45%) 1 year: 80% 5 years 42% NR Yes, neoadjuvant,


(2022)161 patients (71%) 3 years: 63% sometimes com-
had tumours bined with loco­
5 years: 49%
larger ≥5 cm regional therapy
5-year OS was 100% in (10 patients received
patients who received neoadjuvant loco­
both neoadjuvant chemo­ regional therapy
therapy and locoregional with or without
therapy, compared with neoadjuvant chemo-
77%, 57% and 41% at 1, 3 therapy); 11 patients
and 5 years, respectively received adjuvant
in those who did not or chemotherapy
received neoadjuvant
locoregional therapy
alone

Nature Reviews Clinical Oncology | Volume 20 | July 2023 | 470–486 481


Review article

Table 3 (continued) | Summary of key contemporary studies of liver transplantation in patients with iCCA

Study (year of Tumour size iCCAs discovered Patients OS RFS Recurrence rate Chemotherapy
publication) (n) incidentally/initially with treatment
misdiagnosed as HCC cirrhosis (n)

Retrospective studies in patients with large multifocal iCCAs diagnosed prior to liver transplantation (continued)
McMillan et al. Among 18 NA 6 (33%) 1 year: 100% 3 years: 52% 7 (38.9%); median Yes, neoadjuvant
(2022)152 patients: 3 years: 71% time to recurrence and adjuvant
median 11 months (range
5 years: 57%
number of 4–42 months)
tumours 2
(1–11); median
tumour size
10.4 cm
(2.5–19.9 cm)
Ongoing prospective clinical trials
University Single iCCA NA 100% NR (5-year OS is primary NR NR (secondary end NR
Health Network ≤2 cm with no end point) (secondary point)
Toronto phase II extrahepatic end point)
multicentre trial disease or
NCT02878473 vascular
(NA) invasion (target
of 30 patients
who are not
candidates
for tumour
resection and
have serum
CA19.9 levels
≤ 100 ng/ml)
Oslo University NA; NA NR NR (3-year OS is primary NR NR Patients must have
phase II histologically end point) (secondary stable disease
TESLA trial proven iCCA end point) after ≥6 months
with no of systemic
NCT04556214
extrahepatic chemotherapy
(NA)
disease, or locoregional
vascular treatment
invasion or
lymph node
involvement
(target of
15 patients
who are not
candidates
for tumour
resection)
University NA; NA NR NR (5-year OS is primary NR (5-year NR Patients must have
Health Network histologically end point; 1-year OS is RFS is stable disease or
Toronto phase II proven iCCA secondary end point) secondary tumour regression
single-centre with no end point) lasting ≥6 months
trial extrahepatic after systemic
disease, chemotherapy
NCT04195503
vascular or locoregional
(NA)
invasion or treatment
lymph node
involvement
(target of
10 patients
who are not
candidates
for tumour
resection
and have a
potential live
liver donor)
HCC, hepatocellular carcinoma; iCCA, intrahepatic cholangiocarcinoma; IQR, interquartile range; NA, not applicable; NR, not reported; OS, overall survival; RFS, recurrence-free survival.
a
Data for patients with iCCA alone (n = 8), as opposed to concurrent iCCA and HCC (n = 7) or mixed HCC–iCCA (n = 1). bData for patients with iCCA and mixed HCC–iCCA tumours >2 cm but
≤5 cm (n = 24). cData for patients with iCCA ≤2 cm and mixed HCC–iCCA tumours (n = 25).

Nature Reviews Clinical Oncology | Volume 20 | July 2023 | 470–486 482


Review article

patients for transplantation and finding an allograft. Before liver trans- of response owing to acquired resistance; combinatorial approaches
plantation can be recommended for such patients outside clinical will need to be examined in model systems and patients in order to
trials, several questions remain to be answered. First, do patients with a improve outcomes. The use of neoadjuvant and adjuvant therapies in
known diagnosis of iCCA have similar outcomes to those in whom iCCA combination with surgery and/or locoregional therapies to improve
was diagnosed incidentally? Second, should these patients undergo outcomes is also an unmet need that warrants further studies. Finally,
neoadjuvant treatments? Third, should this transplant indication be selection of patients who will benefit from liver transplantation will
limited to single iCCAs of ≤2 cm or could this be expanded to those require national organ allocation systems to acknowledge patients
with single larger tumours? with iCCA as candidates for this procedure. We look forward to future
With advances in the understanding of iCCA biology and improve- updates on CCA, which will hopefully report solutions to these unmet
ments in patient management, the concept of considering liver trans- needs in patients with this disease.
plantation has also been revisited for patients with unresectable iCCA,
no extrahepatic disease and a sustained response to systemic and/or Published online: 15 May 2023
local therapies18. The largest prospective study to date involved a series
References
of 65 patients referred for liver transplantation evaluation; 37 patients 1. Banales, J. M. et al. Cholangiocarcinoma: state-of-the-art knowledge and challenges.
met the criteria and were listed, although five patients were subse- Liver Int. 39, 5–6 (2019).
2. Brindley, P. J. et al. Cholangiocarcinoma. Nat. Rev. Dis. Prim. 7, 65 (2021).
quently excluded owing to conversion to resectable disease with the
3. Banales, J. M. et al. Cholangiocarcinoma 2020: the next horizon in mechanisms and
stipulated neoadjuvant chemotherapy, and 18 eventually received management. Nat. Rev. Gastroenterol. Hepatol. 17, 557–588 (2020).
a liver transplant152. The results of that study demonstrated a 5-year 4. Rizvi, S., Khan, S. A., Hallemeier, C. L., Kelley, R. K. & Gores, G. J.
Cholangiocarcinoma– evolving concepts and therapeutic strategies. Nat. Rev. Clin.
OS of 57% and a 3-year disease-free survival of 52% after transplanta-
Oncol. 15, 95–111 (2018).
tion152. Several questions remain, and these results need to be externally 5. Affo, S. et al. Promotion of cholangiocarcinoma growth by diverse cancer-associated
validated, although they are encouraging and will serve as a baseline in fibroblast subpopulations. Cancer Cell 39, 883 (2021).
6. Yang, J. D. et al. DNA methylation markers for detection of cholangiocarcinoma: discovery,
future studies to identify the optimal candidates for liver transplanta-
validation, and clinical testing in biliary brushings and plasma. Hepatol. Commun. 5,
tion153. To our knowledge, several centres are now considering liver 1448–1459 (2021).
transplantation as an option for patients with unresectable locally 7. Zill, O. A. et al. Cell-free DNA next-generation sequencing in pancreatobiliary
carcinomas. Cancer Discov. 5, 1040–1048 (2015).
advanced iCCA who have a sustained response to systemic and/or local
8. Wu, M. J., Shi, L., Merritt, J., Zhu, A. X. & Bardeesy, N. Biology of IDH mutant
therapies and no evidence of extrahepatic disease at any time point. cholangiocarcinoma. Hepatology 75, 1322–1337 (2022).
With the development of molecularly targeted therapies for iCCA and a 9. Kelley, R. K., Bridgewater, J., Gores, G. J. & Zhu, A. X. Systemic therapies for intrahepatic
cholangiocarcinoma. J. Hepatol. 72, 353–363 (2020).
better understanding of mutations associated with a favourable prog- 10. Abou-Alfa, G. K. et al. Pemigatinib for previously treated, locally advanced or metastatic
nosis, such as FGFR genetic aberrations154, the proportion of patients cholangiocarcinoma: a multicentre, open-label, phase 2 study. Lancet Oncol. 21,
who might be eligible for liver transplantation in the future might 671–684 (2020).
11. Javle, M. et al. Infigratinib (BGJ398) in previously treated patients with advanced or
increase. When considering alternative treatments for these patients, metastatic cholangiocarcinoma with FGFR2 fusions or rearrangements: mature results
the options are extremely limited owing to very poor outcomes with from a multicentre, open-label, single-arm, phase 2 study. Lancet Gastroenterol. Hepatol.
resection or hepatic artery infusion pump chemotherapy155. 6, 803–815 (2021).
12. Goyal, L. et al. Futibatinib for FGFR2-rearranged intrahepatic cholangiocarcinoma.
In the next few years, more data on liver transplantation for iCCA N. Engl. J. Med. 388, 228–239 (2023).
will be published and this treatment modality is expected to become 13. US Food and Drug Administration. FDA grants accelerated approval to pemigatinib for
available to patients at many centres. Until then, liver transplantation cholangiocarcinoma with an FGFR2 rearrangement or fusion. FDA https://www.fda.
gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-
should be performed under strict research protocols with stringent pemigatinib-cholangiocarcinoma-fgfr2-rearrangement-or-fusion (2020).
inclusion criteria. 14. US Food and Drug Administration. FDA grants accelerated approval to infigratinib
for metastatic cholangiocarcinoma. FDA https://www.fda.gov/drugs/resources-

Conclusions information-approved-drugs/fda-grants-accelerated-approval-infigratinib-metastatic-
cholangiocarcinoma (2021).
As is evident from this update, many crucial scientific and clinical ques- 15. US Food and Drug Administration. FDA grants accelerated approval to futibatinib for
tions remain despite the recent progress in CCA research and manage- cholangiocarcinoma. FDA https://www.fda.gov/drugs/resources-information-approved-
drugs/fda-grants-accelerated-approval-futibatinib-cholangiocarcinoma (2022).
ment. Although the TIME presents potential targets for the treatment 16. Abou-Alfa, G. K. et al. Ivosidenib in IDH1-mutant, chemotherapy-refractory
of CCA, the limited antitumour activity of ICIs in this disease highlights cholangiocarcinoma (ClarIDHy): a multicentre, randomised, double-blind, placebo-
the need for a more in-depth and fundamental understanding of the controlled, phase 3 study. Lancet Oncol. 21, 796–807 (2020).
17. US Food and Drug Administration. FDA approves ivosidenib for advanced or metastatic
immunosuppressive cell populations. Further information will also cholangiocarcinoma. FDA https://www.fda.gov/drugs/resources-information-approved-
be needed to stratify TIME-based subgroups for a precision therapy drugs/fda-approves-ivosidenib-advanced-or-metastatic-cholangiocarcinoma (2022).
18. Gravely, A. K., Vibert, E. & Sapisochin, G. Surgical treatment of intrahepatic
approach, and perhaps some subtypes such as the immune-desert
cholangiocarcinoma. J. Hepatol. https://doi.org/10.1016/j.jhep.2022.01.004 (2022).
tumours will prove difficult if not impossible to target effectively. Given 19. Wang, J., Loeuillard, E., Gores, G. J. & Ilyas, S. I. Cholangiocarcinoma: what are the most
the heterogeneity of CAFs in CCAs, targeting this cell popu­lation is valuable therapeutic targets – cancer-associated fibroblasts, immune cells, or beyond
T cells? Expert. Opin. Ther. Targets 25, 835–845 (2021).
also likely to prove more difficult than originally conceptualized.
20. Loeuillard, E. et al. Targeting tumor-associated macrophages and granulocytic myeloid-
Better diagnostic modalities are necessary for pCCA and perhaps the derived suppressor cells augments PD-1 blockade in cholangiocarcinoma. J. Clin. Invest.
emerging technology of measuring cfDNA and ctDNA will address this 130, 5380–5396 (2020).
21. Ruffolo, L. I. et al. GM-CSF drives myelopoiesis, recruitment and polarisation of
problem; the use of such assays would also greatly facilitate monitor-
tumour-associated macrophages in cholangiocarcinoma and systemic blockade
ing of disease activity following surgical resection or during systemic facilitates antitumour immunity. Gut 71, 1386–1398 (2022).
therapy. Indeed, we are optimistic that these liquid biopsy assays will 22. Zhang, Q. et al. Gut microbiome directs hepatocytes to recruit MDSCs and promote
cholangiocarcinoma. Cancer Discov. 11, 1248–1267 (2021).
achieve clinical utility in the management of CCA. Although an impor-
23. Gani, F. et al. Program death 1 immune checkpoint and tumor microenvironment:
tant advance, targeted therapies for patients with FGFR2 fusions or IDH1 implications for patients with intrahepatic cholangiocarcinoma. Ann. Surg. Oncol. 23,
mutations are often met with intrinsic resistance or a short durability 2610–2617 (2016).

Nature Reviews Clinical Oncology | Volume 20 | July 2023 | 470–486 483


Review article

24. Zhou, G. et al. Reduction of immunosuppressive tumor microenvironment in 57. Martin-Serrano, M. A. et al. Novel microenvironment-based classification of intrahepatic
cholangiocarcinoma by ex vivo targeting immune checkpoint molecules. J. Hepatol. 71, cholangiocarcinoma with therapeutic implications. Gut https://doi.org/10.1136/
753–762 (2019). gutjnl-2021-326514 (2022).
25. Ghidini, M. et al. Characterisation of the immune-related transcriptome in resected 58. Zhang, M. et al. Single-cell transcriptomic architecture and intercellular crosstalk of
biliary tract cancers. Eur. J. Cancer 86, 158–165 (2017). human intrahepatic cholangiocarcinoma. J. Hepatol. 73, 1118–1130 (2020).
26. Ma, L. et al. Tumor cell biodiversity drives microenvironmental reprogramming in liver 59. Ravichandra, A., Bhattacharjee, S. & Affo, S. Cancer-associated fibroblasts in intrahepatic
cancer. Cancer Cell 36, 418–430.e6 (2019). cholangiocarcinoma progression and therapeutic resistance. Adv. Cancer Res. 156,
27. Fabris, L., Sato, K., Alpini, G. & Strazzabosco, M. The tumor microenvironment in 201–226 (2022).
cholangiocarcinoma progression. Hepatology 73, 75–85 (2021). 60. Chen, Y. et al. Type I collagen deletion in αSMA+ myofibroblasts augments immune
28. Piha-Paul, S. A. et al. Efficacy and safety of pembrolizumab for the treatment of advanced suppression and accelerates progression of pancreatic cancer. Cancer Cell 39,
biliary cancer: results from the KEYNOTE-158 and KEYNOTE-028 studies. Int. J. Cancer 548–565.e6 (2021).
147, 2190–2198 (2020). 61. Liu, M. C. et al. Sensitive and specific multi-cancer detection and localization using
29. Buettner, S. et al. The impact of neutrophil-to-lymphocyte ratio and platelet-to- methylation signatures in cell-free DNA. Ann. Oncol. 31, 745–759 (2020).
lymphocyte ratio among patients with intrahepatic cholangiocarcinoma. Surgery 164, 62. Cohen, J. D. et al. Detection and localization of surgically resectable cancers with a
411–418 (2018). multi-analyte blood test. Science 359, 926–930 (2018).
30. Peng, D. et al. Lymphocyte to monocyte ratio predicts resectability and early recurrence 63. Wasenang, W., Chaiyarit, P., Proungvitaya, S. & Limpaiboon, T. Serum cell-free DNA
of Bismuth-Corlette type IV hilar cholangiocarcinoma. J. Gastrointest. Surg. 24, 330–340 methylation of OPCML and HOXD9 as a biomarker that may aid in differential diagnosis
(2020). between cholangiocarcinoma and other biliary diseases. Clin. Epigenetics 11, 39
31. Tsilimigras, D. I. et al. The systemic immune-inflammation index predicts prognosis in (2019).
intrahepatic cholangiocarcinoma: an international multi-institutional analysis. HPB 22, 64. Ettrich, T. J. et al. Genotyping of circulating tumor DNA in cholangiocarcinoma reveals
1667–1674 (2020). diagnostic and prognostic information. Sci. Rep. 9, 13261 (2019).
32. Sun, D. et al. CD86+/CD206+ tumor-associated macrophages predict prognosis of 65. Mody, K. et al. Circulating tumor DNA profiling of advanced biliary tract cancers.
patients with intrahepatic cholangiocarcinoma. PeerJ 8, e8458 (2020). JCO Precis. Oncol. 3, 1–9 (2019).
33. Yuan, D. et al. Kupffer cell-derived TNF triggers cholangiocellular tumorigenesis through 66. Moreno Luna, L. E. et al. Advanced cytologic techniques for the detection of malignant
JNK due to chronic mitochondrial dysfunction and ROS. Cancer Cell 31, 771–789.e6 pancreatobiliary strictures. Gastroenterology 131, 1064–1072 (2006).
(2017). 67. Barr Fritcher, E. G. et al. An optimized set of fluorescence in situ hybridization probes for
34. Boulter, L. et al. WNT signaling drives cholangiocarcinoma growth and can be detection of pancreatobiliary tract cancer in cytology brush samples. Gastroenterology
pharmacologically inhibited. J. Clin. Invest. 125, 1269–1285 (2015). 149, 1813–1824.e1 (2015).
35. Dwyer, B. J. et al. TWEAK/Fn14 signalling promotes cholangiocarcinoma niche formation 68. Gonda, T. A. et al. Mutation profile and fluorescence in situ hybridization analyses
and progression. J. Hepatol. 74, 860–872 (2021). increase detection of malignancies in biliary strictures. Clin. Gastroenterol. Hepatol. 15,
36. Veglia, F., Perego, M. & Gabrilovich, D. Myeloid-derived suppressor cells coming of age. 913–919.e1 (2017).
Nat. Immunol. 19, 108–119 (2018). 69. Singhi, A. D. et al. Integrating next-generation sequencing to endoscopic retrograde
37. Xu, X. D. et al. Circulating myeloid-derived suppressor cells in patients with pancreatic cholangiopancreatography (ERCP)-obtained biliary specimens improves the detection
cancer. Hepatobiliary Pancreat. Dis. Int. 15, 99–105 (2016). and management of patients with malignant bile duct strictures. Gut 69, 52–61 (2020).
38. Meyer, C. et al. Frequencies of circulating MDSC correlate with clinical outcome of 70. Arechederra, M. et al. Next-generation sequencing of bile cell-free DNA for the early
melanoma patients treated with ipilimumab. Cancer Immunol. Immunother. 63, 247–257 detection of patients with malignant biliary strictures. Gut 71, 1141–1151 (2022).
(2014). 71. Andresen, K. et al. Four DNA methylation biomarkers in biliary brush samples accurately
39. Song, G. et al. Single-cell transcriptomic analysis suggests two molecularly subtypes identify the presence of cholangiocarcinoma. Hepatology 61, 1651–1659 (2015).
of intrahepatic cholangiocarcinoma. Nat. Commun. 13, 1642 (2022). 72. Loi, E. et al. HOXD8 hypermethylation as a fully sensitive and specific biomarker for
40. Bao, X. et al. Molecular subgroups of intrahepatic cholangiocarcinoma discovered by biliary tract cancer detectable in tissue and bile samples. Br. J. Cancer 126, 1783–1794
single-cell RNA sequencing-assisted multiomics analysis. Cancer Immunol. Res. 10, (2022).
811–828 (2022). 73. Shigehara, K. et al. Real-time PCR-based analysis of the human bile microRNAome
41. Keenan, B. P. et al. Circulating monocytes associated with anti-PD-1 resistance in human identifies miR-9 as a potential diagnostic biomarker for biliary tract cancer. PLoS ONE 6,
biliary cancer induce T cell paralysis. Cell Rep. 40, 111384 (2022). e23584 (2011).
42. Tavazoie, M. F. et al. LXR/ApoE activation restricts innate immune suppression in cancer. 74. Han, H. S. et al. Bile-derived circulating extracellular miR-30d-5p and miR-92a-3p as
Cell 172, 825–840.e18 (2018). potential biomarkers for cholangiocarcinoma. Hepatobiliary Pancreat. Dis. Int. 19, 41–50
43. Vonderheide, R. H. CD40 agonist antibodies in cancer immunotherapy. Annu. Rev. Med. (2020).
71, 47–58 (2020). 75. Berchuck, J. E. et al. The clinical landscape of cell-free DNA alterations in 1671 patients
44. Diggs, L. P. et al. CD40-mediated immune cell activation enhances response to with advanced biliary tract cancer. Ann. Oncol. 33, 1269–1283 (2022).
anti-PD-1 in murine intrahepatic cholangiocarcinoma. J. Hepatol. 74, 1145–1154 76. Andersen, J. B. et al. Genomic and genetic characterization of cholangiocarcinoma
(2021). identifies therapeutic targets for tyrosine kinase inhibitors. Gastroenterology 142,
45. Wabitsch, S. et al. Anti-PD-1 in combination with trametinib suppresses tumor growth and 1021–1031.e15 (2012).
improves survival of intrahepatic cholangiocarcinoma in mice. Cell Mol. Gastroenterol. 77. Montal, R. et al. Molecular classification and therapeutic targets in extrahepatic
Hepatol. 12, 1166–1178 (2021). cholangiocarcinoma. J. Hepatol. 73, 315–327 (2020).
46. Affo, S., Yu, L. X. & Schwabe, R. F. The role of cancer-associated fibroblasts and fibrosis 78. Sia, D. et al. Integrative molecular analysis of intrahepatic cholangiocarcinoma reveals 2
in liver cancer. Annu. Rev. Pathol. 12, 153–186 (2017). classes that have different outcomes. Gastroenterology 144, 829–840 (2013).
47. Brivio, S., Cadamuro, M., Strazzabosco, M. & Fabris, L. Tumor reactive stroma in 79. Job, S. et al. Identification of four immune subtypes characterized by distinct
cholangiocarcinoma: the fuel behind cancer aggressiveness. World J. Hepatol. 9, composition and functions of tumor microenvironment in intrahepatic
455–468 (2017). cholangiocarcinoma. Hepatology 72, 965–981 (2020).
48. Sirica, A. E., Campbell, D. J. & Dumur, C. I. Cancer-associated fibroblasts in intrahepatic 80. Lowery, M. A. et al. Comprehensive molecular profiling of intrahepatic and extrahepatic
cholangiocarcinoma. Curr. Opin. Gastroenterol. 27, 276–284 (2011). cholangiocarcinomas: potential targets for intervention. Clin. Cancer Res. 24, 4154–4161
49. Sirica, A. E. & Gores, G. J. Desmoplastic stroma and cholangiocarcinoma: clinical (2018).
implications and therapeutic targeting. Hepatology 59, 2397–2402 (2014). 81. Weinberg, B. A. et al. Molecular profiling of biliary cancers reveals distinct molecular
50. Fabris, L. et al. The tumour microenvironment and immune milieu of alterations and potential therapeutic targets. J. Gastrointest. Oncol. 10, 652–662 (2019).
cholangiocarcinoma. Liver Int. 39, 63–78 (2019). 82. Wardell, C. P. et al. Genomic characterization of biliary tract cancers identifies driver
51. Mertens, J. C. et al. Therapeutic effects of deleting cancer-associated fibroblasts genes and predisposing mutations. J. Hepatol. 68, 959–969 (2018).
in cholangiocarcinoma. Cancer Res. 73, 897–907 (2013). 83. Jusakul, A. et al. Whole-genome and epigenomic landscapes of etiologically distinct
52. Chen, Y., McAndrews, K. M. & Kalluri, R. Clinical and therapeutic relevance of subtypes of cholangiocarcinoma. Cancer Discov. 7, 1116–1135 (2017).
cancer-associated fibroblasts. Nat. Rev. Clin. Oncol. 18, 792–804 (2021). 84. Nakamura, H. et al. Genomic spectra of biliary tract cancer. Nat. Genet. 47, 1003–1010
53. Campbell, D. J., Dumur, C. I., Lamour, N. F., Dewitt, J. L. & Sirica, A. E. Novel organotypic (2015).
culture model of cholangiocarcinoma progression. Hepatol. Res. 42, 1119–1130 85. Boscoe, A. N., Rolland, C. & Kelley, R. K. Frequency and prognostic significance of
(2012). isocitrate dehydrogenase 1 mutations in cholangiocarcinoma: a systematic literature
54. Biffi, G. & Tuveson, D. A. Diversity and biology of cancer-associated fibroblasts. Physiol. review. J. Gastrointest. Oncol. 10, 751–765 (2019).
Rev. 101, 147–176 (2021). 86. Chaisaingmongkol, J. et al. Common molecular subtypes among Asian hepatocellular
55. Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 16, 582–598 carcinoma and cholangiocarcinoma. Cancer Cell 32, 57–70.e3 (2017).
(2016). 87. Chan-On, W. et al. Exome sequencing identifies distinct mutational patterns in liver
56. Bhattacharjee, S. et al. Tumor restriction by type I collagen opposes tumor-promoting fluke-related and non-infection-related bile duct cancers. Nat. Genet. 45, 1474–1478 (2013).
effects of cancer-associated fibroblasts. J. Clin. Invest. https://doi.org/10.1172/JCI146987 88. Bekaii-Saab, T. S., Bridgewater, J. & Normanno, N. Practical considerations in screening
(2021). for genetic alterations in cholangiocarcinoma. Ann. Oncol. 32, 1111–1126 (2021).

Nature Reviews Clinical Oncology | Volume 20 | July 2023 | 470–486 484


Review article

89. Lamarca, A. et al. Molecular profiling in daily clinical practice: practicalities in advanced 118. Harding, J. J. et al. Targeting HER2 mutation-positive advanced biliary tract cancers
cholangiocarcinoma and other biliary tract cancers. J. Clin. Med. https://doi.org/10.3390/ with neratinib: final results from the phase 2 SUMMIT basket trial. J. Clin. Oncol. 40,
jcm9092854 (2020). 4079–4079 (2022).
90. National Comprehensive Cancer Network. NCCN guidelines: hepatobiliary cancer. 119. Meric-Bernstam, F. et al. Zanidatamab (ZW25) in HER2-positive biliary tract cancers
NCCN https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1438 (2022). (BTCs): results from a phase I study [abstract]. J. Clin. Oncol. 39 (Suppl. 3), 299 (2021).
91. Haugsten, E. M., Wiedlocha, A., Olsnes, S. & Wesche, J. Roles of fibroblast growth factor 120. Westphalen, C. B. et al. Genomic context of NTRK1/2/3 fusion-positive tumours from a
receptors in carcinogenesis. Mol. Cancer Res. 8, 1439–1452 (2010). large real-world population. NPJ Precis. Oncol. 5, 69 (2021).
92. Babina, I. S. & Turner, N. C. Advances and challenges in targeting FGFR signalling 121. Drilon, A. et al. Efficacy of larotrectinib in TRK fusion-positive cancers in adults and
in cancer. Nat. Rev. Cancer 17, 318–332 (2017). children. N. Engl. J. Med. 378, 731–739 (2018).
93. Wu, Y. M. et al. Identification of targetable FGFR gene fusions in diverse cancers. 122. US Food and Drug Administration. FDA approves larotrectinib for solid tumors with
Cancer Discov. 3, 636–647 (2013). NTRK gene fusions. FDA https://www.fda.gov/drugs/fda-approves-larotrectinib-solid-
94. Graham, R. P. et al. Fibroblast growth factor receptor 2 translocations in intrahepatic tumors-ntrk-gene-fusions-0 (2018).
cholangiocarcinoma. Hum. Pathol. 45, 1630–1638 (2014). 123. Doebele, R. C. et al. Entrectinib in patients with advanced or metastatic NTRK fusion-
95. Arai, Y. et al. Fibroblast growth factor receptor 2 tyrosine kinase fusions define a unique positive solid tumours: integrated analysis of three phase 1-2 trials. Lancet Oncol. 21,
molecular subtype of cholangiocarcinoma. Hepatology 59, 1427–1434 (2014). 271–282 (2020).
96. Becker, Z. BridgeBio says it’s undeterred after Truseltiq partner withdraws application, 124. US Food and Drug Administration. FDA approves entrectinib for NTRK solid tumors and
discontinues drug. FIERCE Pharma https://www.fiercepharma.com/pharma/bridgebios- ROS-1 NSCLC. FDA https://www.fda.gov/drugs/resources-information-approved-drugs/
cancer-drug-truseltiqs-future-unclear-partner-helsinn-withdrawing-nda (2022). fda-approves-entrectinib-ntrk-solid-tumors-and-ros-1-nsclc (2019).
97. Droz Dit Busset, M. et al. Derazantinib for patients with intrahepatic cholangiocarcinoma 125. Parimi, V. et al. Genomic landscape of 891 RET fusions detected across diverse solid
harboring FGFR2 fusions/rearrangements: primary results from the phase II study tumor types. NPJ Precis. Oncol. 7, 10 (2023).
FIDES-01 [abstract 47P]. Ann. Oncol. 32 (Suppl. 5), S376 (2021). 126. Subbiah, V. et al. Pan-cancer efficacy of pralsetinib in patients with RET fusion-positive
98. Krook, M. A. et al. Efficacy of FGFR inhibitors and combination therapies for acquired solid tumors from the phase 1/2 ARROW trial. Nat. Med. 28, 1640–1645 (2022).
resistance in FGFR2-fusion cholangiocarcinoma. Mol. Cancer Ther. 19, 847–857 (2020). 127. Subbiah, V. et al. Tumour-agnostic efficacy and safety of selpercatinib in patients with
99. Krook, M. A. et al. Tumor heterogeneity and acquired drug resistance in FGFR2-fusion- RET fusion-positive solid tumours other than lung or thyroid tumours (LIBRETTO-001):
positive cholangiocarcinoma through rapid research autopsy. Cold Spring Harb. Mol. a phase 1/2, open-label, basket trial. Lancet Oncol. 23, 1261–1273 (2022).
Case Stud. https://doi.org/10.1101/mcs.a004002 (2019). 128. Valle, J. W., Kelley, R. K., Nervi, B., Oh, D. Y. & Zhu, A. X. Biliary tract cancer. Lancet 397,
100. Goyal, L. et al. Polyclonal secondary FGFR2 mutations drive acquired resistance to FGFR 428–444 (2021).
inhibition in patients with FGFR2 fusion-positive cholangiocarcinoma. Cancer Discov. 7, 129. Lamarca, A., Edeline, J. & Goyal, L. How I treat biliary tract cancer. ESMO Open 7, 100378
252–263 (2017). (2022).
101. Goyal, L. et al. TAS-120 overcomes resistance to ATP-competitive FGFR inhibitors in 130. Le, D. T. et al. Mismatch repair deficiency predicts response of solid tumors to PD-1
patients with FGFR2 fusion-positive intrahepatic cholangiocarcinoma. Cancer Discov. 9, blockade. Science 357, 409–413 (2017).
1064–1079 (2019). 131. Maio, M. et al. Pembrolizumab in microsatellite instability high or mismatch repair
102. Sootome, H. et al. Futibatinib is a novel irreversible FGFR 1-4 inhibitor that shows deficient cancers: updated analysis from the phase 2 KEYNOTE-158 study. Ann. Oncol.
selective antitumor activity against FGFR-deregulated tumors. Cancer Res. 80, https://doi.org/10.1016/j.annonc.2022.05.519 (2022).
4986–4997 (2020). 132. US Food and Drug Administration. FDA grants accelerated approval to pembrolizumab
103. Meric-Bernstam, F. et al. Futibatinib, an irreversible FGFR1-4 inhibitor, in patients with for first tissue/site agnostic indication. FDA https://www.fda.gov/drugs/resources-
advanced solid tumors harboring FGF/FGFR aberrations: a phase I dose-expansion study. information-approved-drugs/fda-grants-accelerated-approval-pembrolizumab-
Cancer Discov. 12, 402–415 (2022). first-tissuesite-agnostic-indication (2017).
104. Casaletto, J. et al. RLY-4008, a novel precision therapy for FGFR2-driven cancers 133. US Food and Drug Administration. FDA approves pembrolizumab for adults and
designed to potently and selectively inhibit FGFR2 and FGFR2 resistance mutations children with TMB-H solid tumors. FDA https://www.fda.gov/drugs/drug-approvals-
[abstract]. Cancer Res. 81 (Suppl. 13), 1455 (2021). and-databases/fda-approves-pembrolizumab-adults-and-children-tmb-h-solid-tumors
105. Franovic, A. et al. Activity of KIN-3248, a next-generation pan-FGFR inhibitor, against (2020).
acquired FGFR-gatekeeper and molecular-brake drug resistance mutations [abstract]. 134. Marabelle, A. et al. Association of tumour mutational burden with outcomes in patients
J. Clin. Oncol. 40 (Suppl. 4), 461 (2022). with advanced solid tumours treated with pembrolizumab: prospective biomarker
106. Goyal, L. et al. First results of RLY-4008, a potent and highly selective FGFR2 inhibitor in analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol. 21,
a first-in-human study in patients with FGFR2-altered cholangiocarcinoma and multiple 1353–1365 (2020).
solid tumors [abstract]. Mol. Cancer Ther. 20 (Suppl. 12), P02-02 (2021). 135. Ma, K. et al. PD-L1 and PD-1 expression correlate with prognosis in extrahepatic
107. Lu, C. & Thompson, C. B. Metabolic regulation of epigenetics. Cell Metab. 16, 9–17 (2012). cholangiocarcinoma. Oncol. Lett. 14, 250–256 (2017).
108. Inoue, S. et al. Mutant IDH1 downregulates ATM and alters DNA repair and sensitivity to 136. Zhu, Y. et al. Programmed death ligand 1 expression in human intrahepatic
DNA damage independent of TET2. Cancer Cell 30, 337–348 (2016). cholangiocarcinoma and its association with prognosis and CD8+ T-cell immune
109. Sulkowski, P. L. et al. 2-Hydroxyglutarate produced by neomorphic IDH mutations responses. Cancer Manag. Res. 10, 4113–4123 (2018).
suppresses homologous recombination and induces PARP inhibitor sensitivity. 137. Fluxa, P. et al. High CD8+ and absence of Foxp3+ T lymphocytes infiltration in gallbladder
Sci. Transl. Med. https://doi.org/10.1126/scitranslmed.aal2463 (2017). tumors correlate with prolonged patients survival. BMC Cancer 18, 243 (2018).
110. Valle, J. W., Lamarca, A., Goyal, L., Barriuso, J. & Zhu, A. X. New horizons for precision 138. Goeppert, B. et al. Major histocompatibility complex class I expression impacts on
medicine in biliary tract cancers. Cancer Discov. 7, 943–962 (2017). patient survival and type and density of immune cells in biliary tract cancer. Br. J. Cancer
111. Subbiah, V. et al. Dabrafenib plus trametinib in patients with BRAFV600E-mutated biliary 113, 1343–1349 (2015).
tract cancer (ROAR): a phase 2, open-label, single-arm, multicentre basket trial. 139. Sun, D. et al. Anti-PD-1 therapy combined with chemotherapy in patients with advanced
Lancet Oncol. 21, 1234–1243 (2020). biliary tract cancer. Cancer Immunol. Immunother. 68, 1527–1535 (2019).
112. US Food and Drug Administration. FDA grants accelerated approval of dabrafenib in 140. Sahai, V. et al. A multicenter randomized phase II study of nivolumab in combination
combination with trametinib for unresectable or metastatic solid tumors with BRAF with gemcitabine/cisplatin or ipilimumab as first-line therapy for patients with advanced
V600E mutation. FDA https://www.fda.gov/drugs/resources-information-approved-drugs/ unresectable biliary tract cancer (BilT-01) [abstract]. J. Clin. Oncol. 38 (Suppl. 15), 4582
fda-grants-accelerated-approval-dabrafenib-combination-trametinib-unresectable-or- (2020).
metastatic-solid (2022). 141. Liu, T. et al. Toripalimab with chemotherapy as first-line treatment for advanced biliary
113. Subbiah, V. et al. Pan-cancer efficacy of vemurafenib in BRAFV600-mutant non-melanoma tract tumors: a preliminary analysis of safety and efficacy of an open-label phase II
cancers. Cancer Discov. 10, 657–663 (2020). clinical study [abstract 53P]. Ann. Oncol. 31 (Suppl. 4), S261 (2020).
114. Galdy, S. et al. HER2/HER3 pathway in biliary tract malignancies; systematic review 142. Oh, D. Y. et al. Gemcitabine and cisplatin plus durvalumab with or without tremelimumab
and meta-analysis: a potential therapeutic target? Cancer Metastasis Rev. 36, 141–157 in chemotherapy-naive patients with advanced biliary tract cancer: an open-label,
(2017). single-centre, phase 2 study. Lancet Gastroenterol. Hepatol. 7, 522–532 (2022).
115. Javle, M. et al. Pertuzumab and trastuzumab for HER2-positive, metastatic biliary 143. Oh, D.-Y. et al. Durvalumab plus gemcitabine and cisplatin in advanced biliary tract
tract cancer (MyPathway): a multicentre, open-label, phase 2a, multiple basket study. cancer. NEJM Evid. https://doi.org/10.1056/EVIDoa2200015 (2022).
Lancet Oncol. 22, 1290–1300 (2021). 144. Burris, H. A. et al. Patient-reported outcomes for the phase 3 TOPAZ-1 study of
116. Ohba, A. et al. Trastuzumab deruxtecan (T-DXd; DS-8201) in patients (pts) with HER2- durvalumab plus gemcitabine and cisplatin in advanced biliary tract cancer [abstract].
expressing unresectable or recurrent biliary tract cancer (BTC): an investigator-initiated J. Clin. Oncol. 40 (Suppl. 16), 4070 (2022).
multicenter phase 2 study (HERB trial) [abstract]. J. Clin. Oncol. 40 (Suppl. 16), 4006 145. He, A. et al. Outcomes by primary tumour location in patients with advanced biliary tract
(2022). cancer treated with durvalumab or placebo plus gemcitabine and cisplatin in the phase 3
117. Lee, C. K. et al. Trastuzumab plus FOLFOX for HER2-positive biliary tract cancer refractory TOPAZ-1 study [abstract O-1]. Ann. Oncol. 33 (Suppl. 4), S378 (2022).
to gemcitabine and cisplatin: a multi-institutional phase 2 trial of the Korean Cancer 146. Okusaka, T. et al. Outcomes by disease status in patients with advanced biliary tract
Study Group (KCSG-HB19-14). Lancet Gastroenterol. Hepatol. https://doi.org/10.1016/ cancer treated wtih durvalumab or placebo plus gemcitabine and cisplatin in the
S2468-1253(22)00335-1 (2022). phase 3 TOPAZ-1 study [abstract 93P]. Ann. Oncol. 33 (Suppl. 9), S1471 (2022).

Nature Reviews Clinical Oncology | Volume 20 | July 2023 | 470–486 485


Review article

147. Vogel, A. et al. Regional subgroup analysis of the phase 3 TOPAZ-1 study of durvalumab (ID 100010434) and by the European Union’s Horizon 2020 research and innovation
(D) plus gemcitabine and cisplatin (GC) in advanced biliary tract cancer (BTC) [abstract]. programme under the Marie Skłodowska-Curie grant agreement no. 847648. The work of
J. Clin. Oncol. 40 (Suppl. 16), 4075 (2022). L.G. is supported by American Cancer Society Clinical Scientist Development Grant 134013‐
148. US Food and Drug Administration. FDA D.I.S.C.O. burst edition: FDA approval of Imfinzi CSDG‐19‐163‐01‐TBG and NIH/NCI Gastrointestinal Cancer SPORE P50 CA127003. A.L. has
(durvalumab) for adult patients with locally advanced or metastatic biliary tract cancer. received funding from The Christie Charity and the European Union’s Horizon 2020 research
FDA https://www.fda.gov/drugs/resources-information-approved-drugs/fda-disco-burst- and innovation programme (grant no. 82551, ESCALON). J.D.Y. has received an American
edition-fda-approval-imfinzi-durvalumab-adult-patients-locally-advanced-or (2022). College of Gastroenterology Junior Faculty Development Award and a U.S. Department of
149. Kelley, R. K. et al. Pembrolizumab in combination with gemcitabine and cisplatin Defense Peer Reviewed Cancer Research Program Career Development Award (CA 191051).
compared with gemcitabine and cisplatin alone for patients with advanced biliary tract
cancer (KEYNOTE-966): a randomised, double-blind, placebo-controlled, phase 3 trial. Author contributions
Lancet https://doi.org/10.1016/S0140-6736(23)00727-4 (2023). S.I.I., J.D.Y. and G.J.G. contributed substantially to discussion of the content and reviewed
150. Villaneuva, L. et al. Lenvatinib plus pembrolizumab for patients with previously treated and/or edited the manuscript before submission. All authors contributed to researching
biliary tract cancers in the multicohort phase II LEAP-005 study [abstract]. J. Clin. Oncol. data for the article and writing the manuscript.
39 (Suppl. 3), 321 (2021).
151. Sapisochin, G. et al. Transplant oncology in primary and metastatic liver tumors: Competing interests
principles, evidence, and opportunities. Ann. Surg. 273, 483–493 (2021). S.I.I. serves as an adviser and consultant to AstraZeneca. L.G. has received research
152. McMillan, R. R. et al. Survival following liver transplantation for locally advanced, funding (via her institution) from Adaptimmune, Bayer, Bristol Myers Squibb, Eisai, Eli Lilly,
unresectable intrahepatic cholangiocarcinoma. Am. J. Transpl. 22, 823–832 (2022). Genentech, Incyte, Leap Therapeutics, Loxo Oncology, Macrogenics, Merck, Novartis,
153. Ivanics, T., Toso, C., Ilyas, S. I. & Sapisochin, G. Transplant oncology in locally advanced Nucana, QED Therapeutics, Relay Therapeutics, Servier and Taiho Oncology, and serves
intrahepatic cholangiocarcinoma: one more step on a long road. Am. J. Transpl. 22, as an adviser or consultant to Alentis Therapeutics, AstraZeneca, Black Diamond,
685–686 (2022). Exelixis, Genentech, H3Biomedicine, Incyte, QED Therapeutics, Servier, Sirtex Medical,
154. Jain, A. et al. Cholangiocarcinoma with FGFR genetic aberrations: a unique clinical Taiho Oncology and Transthera Bio. A.L. declares travel and educational support from
phenotype. JCO Precis. Oncol. 2, 1–12 (2018). Advanced Accelerator Applications, Bayer, Delcath, Ipsen, Mylan, Novartis, Pfizer and
155. Franssen, S. et al. Comparison of hepatic arterial infusion pump chemotherapy vs SirtEx; speaker’s honoraria from Advanced Accelerator Applications, AstraZeneca, Eisai,
resection for patients with multifocal intrahepatic cholangiocarcinoma. JAMA Surg. Incyte, Ipsen, Merck, Pfizer, QED Therapeutics and Servier; and advisory or consultancy
https://doi.org/10.1001/jamasurg.2022.1298 (2022). roles with Albireo Pharma, AstraZeneca, Boehringer Ingelheim, Boston Scientific, Eisai,
156. Sapisochin, G. et al. “Very early” intrahepatic cholangiocarcinoma in cirrhotic patients: GENFIT, Ipsen, Nutricia, QED Therapeutics, Roche, Servier and TransThera Biosciences;
should liver transplantation be reconsidered in these patients? Am. J. Transpl. 14, and is a member of the Knowledge Network and NETConnect Initiatives funded by
660–667 (2014). Ipsen. G.S. has received grant support from Roche, and declares consultancy roles with
157. Sapisochin, G. et al. Liver transplantation for “very early” intrahepatic AstraZeneca, Chiesi, Evidera, Integra, Novartis and Roche. S.A., J.D.Y. and G.J.G. declare
cholangiocarcinoma: international retrospective study supporting a prospective no competing interests.
assessment. Hepatology 64, 1178–1188 (2016).
158. Jung, D. H. et al. Clinicopathological features and prognosis of intrahepatic Additional information
cholangiocarcinoma after liver transplantation and resection. Ann. Transpl. 22, 42–52 Peer review information Nature Reviews Clinical Oncology thanks C. Berasain, H. Francis,
(2017). T. Greten, and J. Harding for their contribution to the peer review of this work.
159. De Martin, E. et al. Analysis of liver resection versus liver transplantation on outcome of
small intrahepatic cholangiocarcinoma and combined hepatocellular-cholangiocarcinoma Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in
in the setting of cirrhosis. Liver Transpl. 26, 785–798 (2020). published maps and institutional affiliations.
160. Lunsford, K. E. et al. Liver transplantation for locally advanced intrahepatic Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to
cholangiocarcinoma treated with neoadjuvant therapy: a prospective case-series. this article under a publishing agreement with the author(s) or other rightsholder(s); author
Lancet Gastroenterol. Hepatol. 3, 337–348 (2018). self-archiving of the accepted manuscript version of this article is solely governed by the
161. Ito, T. et al. A 3-decade, single-center experience of liver transplantation for terms of such publishing agreement and applicable law.
cholangiocarcinoma: impact of era, tumor size, location, and neoadjuvant therapy.
Liver Transpl. 28, 386–396 (2022).
Related links
Acknowledgements ClinicalTrials.gov: https://clinicaltrials.gov/
The work of the authors is supported by the US National Institutes of Health (NIH)/National
Cancer Institute (NCI) grants SPORE P50 CA210964 (to S.I.I. and G.J.G.) and 1K08CA236874 © Springer Nature Limited 2023
(to S.I.I.). The work of S.A. is supported by a fellowship from “la Caixa” Foundation

Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA. 2Department of Immunology, Mayo Clinic, Rochester, MN, USA.
1

3
Liver, Digestive System and Metabolism Research, Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain. 4Department of
Medicine, Mass General Cancer Center, Harvard Medical School, Boston, MA, USA. 5Department of Oncology, OncoHealth Institute, Fundación Jiménez
Díaz University Hospital, Madrid, Spain. 6Department of Medical Oncology, The Christie NHS Foundation, Manchester, UK. 7Division of Cancer Sciences,
University of Manchester, Manchester, UK. 8Ajmera Transplant Program and HPB Surgical Oncology, Toronto General Hospital, University of Toronto,
Toronto, Canada. 9Karsh Division of Gastroenterology and Hepatology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center,
Los Angeles, CA, USA.

Nature Reviews Clinical Oncology | Volume 20 | July 2023 | 470–486 486

You might also like