World Journal of Oncology, ISSN 1920-4531 print, 1920-454X online, Open Access
Article copyright, the authors; Journal compilation copyright, World J Oncol and Elmer Press Inc
Journal website https://wjon.elmerpub.com

Review

Volume 000, Number 000, May 2025, pages 000-000


Immune Checkpoint Inhibition for Hepatocellular Carcinoma, Cholangiocarcinoma, and Combined Hepatocellular-Cholangiocarcinoma

Keiko Shichiria, b, Kaity H. Tunga, c, Kazuaki Takabea, c, d, e, f, g, h, i, David L. Bartlettb

aDepartment of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
bDepartment of Surgery, Allegheny Health Network, Pittsburgh, PA 15212, USA
cDepartment of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences the State University of New York, Buffalo, NY 14203, USA
dDepartment of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
eDepartment of Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
fDepartment of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan
gDepartment of Breast Surgery and Oncology, Tokyo Medical University, Tokyo 160-8402, Japan
hDepartment of Breast Surgery, Fukushima Medical University, Fukushima, Japan
iCorresponding Author: Kazuaki Takabe, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA

Manuscript submitted March 5, 2025, accepted April 26, 2025, published online May 13, 2025
Short title: Immunotherapy for HCC, CCA, and cHCC-CCA
doi: https://doi.org/10.14740/wjon2571

Abstract▴Top 

Combined hepatocellular-cholangiocarcinoma (cHCC-CCA) is a rare primary liver carcinoma that is composed of hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA). Given its low incidence, there is no standardized treatment protocol or systemic regimens. With the development of immune checkpoint inhibitors (ICIs), one of the immunotherapies that modulate the immune system by restoring antitumor immune response, studies have shown promising results for the use of ICI as systemic therapy for advanced solid tumors, including liver cancers. Moreover, prospective clinical studies displayed favorable outcomes of the use of ICIs in HCC and biliary tract cancers. Here, we review the recent evidence in application and comparison of ICIs for HCC, CCA, and cHCC-CCA as well as the future direction of systemic therapy for cHCC-CCA.

Keywords: Combined hepatocellular-cholangiocarcinoma; Hepatocellular carcinoma; Cholangiocarcinoma; Immune checkpoint inhibition

Introduction▴Top 

Combined hepatocellular-cholangiocarcinoma (cHCC-CCA) is a rare primary liver carcinoma with hepatocytic and cholangiocytic differentiations regardless of percentage of each component, diagnosed via routine histochemical stains [1, 2]. Since its first report in 1903, the terminology, definition, and classification of cHCC-CCA have undergone several modifications, likely contributing to its wide reported range of incidence from 0.4% to 14.2% [3-7]. The true incidence is also likely underestimated due to misdiagnosis as hepatocellular carcinoma (HCC) or cholangiocarcinoma (CCA) without tissue diagnosis if patients did not undergo surgical resection. The low incidence and rarity of the disease do not allow for standardization of treatment regimens, and clinicians rely on treatments used for HCC or CCA. Although resection is the only curative treatment, many are found to be unresectable at the time of diagnosis. Recurrence rate is high despite the use of adjuvant therapies. The prognosis of cHCC-CCA is known to be worse than that of HCC, and the 5-year overall survival (OS) is reported to be 20-30% [5, 8-10].

Immunotherapy using immune checkpoint inhibitors (ICIs) is a breakthrough in cancer treatment for many types of cancer [11-13]. Among ICIs, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) blockade and programmed cell death-1 (PD-1) and programmed cell death-ligand 1 (PD-L1) interaction disruption have demonstrated promising clinical outcomes in many cancers, including HCC and biliary tract cancers (BTCs) [14-16]. It is also known that microsatellite instability-high (MSI-H) or mismatch repair deficiency (dMMR) are predictive factors for higher response to ICIs in various cancers [17-19].

Here, we review the recent evidence of ICIs for cHCC-CCA in comparison to that of HCC and CCA.

Pembrolizumab▴Top 

Pembrolizumab is a humanized immunoglobulin G4 monoclonal antibody that binds to the inhibitory immune checkpoint receptor PD-1 expressed on lymphocytes, blocking binding of its ligands, PD-L1 and programmed cell death-ligand 2 (PD-L2), and causes T-cell-mediated tumor destruction [20, 21]. It was first approved for unresectable or metastatic melanoma by the United States Food and Drug Administration (FDA) in 2014 [22] and currently used for various cancers, including MSI-H/dMMR solid tumor [19, 23], non-small cell lung cancer [24], head and neck squamous cell carcinoma [25], gastric cancer [26], urothelial carcinoma [27], and triple-negative breast cancer [12].

Pembrolizumab for HCC

Pembrolizumab was first evaluated as a monotherapy for advanced HCC previously treated with sorafenib in the non-randomized, open-label, phase II KEYNOTE-224 trial, which included 104 patients across 47 medical centers in 10 countries [28]. It demonstrated a 17% objective response rate (ORR) (95% confidence interval (CI): 11-26%). Of the patients, 25% and 1% had a grade 3-4 or grade 5 treatment-related adverse events, respectively [28]. The FDA granted accelerated approval for pembrolizumab for HCC with previous history of sorafenib treatment based on the result of this study. The phase III KEYNOTE-240 trial comparing pembrolizumab to a placebo as second-line treatment for HCC was then conducted, consisting of 413 patients from 119 medical centers in 27 countries [29]. Although this trial did not meet its primary endpoints, OS, progression-free survival (PFS), ORR, and duration of response (DOR) were favorable to pembrolizumab group [29]. The median OS with pembrolizumab versus placebo was 13.9 versus 10.6 months, respectively (hazard ratio (HR): 0.771; 95% CI: 0.617 - 0.964), and the median PFS was 3.0 versus 2.8 months, respectively (HR: 0.718; 95% CI: 0.571 - 0.903). Grade 3 or 4 adverse events were reported for 53.4% of patients on pembrolizumab and for 46.3% of patients on placebo. Pembrolizumab was also assessed in the paralleled phase III KEYNOTE-394 trial in Asian countries, consisting of patients with advanced HCC with progression on or after intolerance to sorafenib or oxaliplatin-based chemotherapy in mainland China, Hong Kong, Republic of Korea, Malaysia, and Taiwan [30]. Pembrolizumab showed statistically significant improvement in OS, PFS, and ORR compared with placebo [30]. Median OS with pembrolizumab or placebo were 14.6 and 13.0 months, respectively (HR for death: 0.79; 95% CI: 0.63 - 0.99; P = 0.0180). Median PFS were 2.6 and 2.3 months, respectively (HR for progression or death: 0.74; 95% CI: 0.60 - 0.92; P = 0.0032). ORR was 12.7% (95% CI: 9.1-17.0%) with pembrolizumab, which was greater than in the placebo group (1.3%, 95% CI: 0.2-4.6%; P < 0.0001). KEYNOTE-240 and KEYNOTE-394 had similar OS HRs (0.78, 95% CI: 0.61 - 1.0; 0.79, 95% CI: 0.63 - 0.99, respectively). PFS HRs were also comparable (KEYNOTE-240 (final analysis): 0.72, 95% CI: 0.57 - 0.90 and KEYNOTE-394 (second interim analysis): 0.74, 95% CI: 0.60 - 0.92). ORR with pembrolizumab versus placebo also demonstrated a similar trend (KEYNOTE-240, 13.8%; KEYNOTE-394, 11.4%) [29, 30].

Pembrolizumab was also evaluated as a first-line monotherapy for patients with advanced HCC from cohort 2 data in phase II KEYNOTE-224 trial [31]. Fifty-one patients were allocated to cohort 2. ORR was 16% (95% CI: 7-29%) with a median DOR of 16 months (range, 3 to 24+ months). Median PFS was 4 months (95% CI: 2 - 8), and an estimated PFS rate at 12 months was 28%. Of the participants, 16% experienced more than grade 3 treatment-related adverse events. The authors concluded that pembrolizumab showed durable antitumor activity, promising OS, and safe profile for patients with advanced HCC with no prior systemic therapy. Currently, National Comprehensive Cancer Network (NCCN) 2024 recommends pembrolizumab as one of other recommended regimens for first-line (category B2) and subsequent-line systemic therapy [32].

Pembrolizumab for CCA

KEYNOTE-158 is a nonrandomized, open-label, multisite phase II study of pembrolizumab monotherapy for MSI-H/dMMR advanced non-colorectal cancer as second-line therapy [18]. A total of 233 patients with MSI-H/dMMR non-colorectal cancer from 18 countries, including 22 (9.4%) patients with CCA, were enrolled. Median follow-up was 13.4 months. Radiographic ORR was 34.3% (95% CI: 28.3-40.8%) with median PFS of 4.1 months (95% CI: 2.4 - 4.9 months). Median OS was 23.5 months (95% CI: 13.5 months - not reached). Of the patients, 14.6% had more than grade 3 treatment-related adverse events. Subgroup analysis of 22 patients with CCA demonstrated an ORR of 40.9% (95% CI: 20.7-63.6%) with a median PFS and OS of 4.2 months (95% CI: 2.1 months - not reached) and 24.3 months (95% CI: 6.5 months - not reached), respectively. In the updated analysis of KEYNOTE-158 with a median follow-up of 37.5 months, 30.8% of 351 patients (95% CI: 25.8-36.2%) achieved an overall response [33]. The median PFS, median OS, and median DOR were 3.5 months (95% CI: 2.3 - 4.2 months), 20.1 months (95% CI: 14.1 - 27.1 months), and 47.5 months (95% CI: 2.1+ months to 51.1+ months), respectively. Subgroup analysis of the same 22 patients showed a median PFS of 4.2 months (95% CI: 2.1 - 24.9 months) and a median OS of 19.4 months (95% CI: 6.5 months - not reached). The ORR was the same as previously reported in KEYNOTE-158. Based on the results of KEYNOTE-158, the FDA approved pembrolizumab monotherapy for patients with unresectable, metastatic MSI-H or dMMR solid tumors that failed previous treatment and lacked alternative treatment options.

The standard first-line regimen for advanced biliary tumor is the combination of gemcitabine and cisplatin, which acts to upregulate the immune system via several mechanisms, such as the recruitment of immune effector cells, enhancement of antigenicity, and downregulation of the immunosuppressive microenvironment [34-36]. The phase III randomized KEYNOTE-966 trial investigated the effect of the combination of pembrolizumab with gemcitabine and cisplatin on advanced BTC by looking at 1,069 patients with unresectable, locally advanced or metastatic BTC, in which 59% had intrahepatic CCA [37]. Of note, eight patients (2%) in the pembrolizumab group and five patients (1%) in the placebo group had cHCC-CCA. In the intention-to-treat population, median OS in the treatment group showed significant improvement compared to the control group (12.7 vs. 10.9 months, HR: 0.83; 95% CI: 0.72 - 0.95; one-sided P = 0.0034) as well as a trend towards improved benefit to add pembrolizumab in patients with intrahepatic CCA. Currently, the NCCN 2024 guideline recommends pembrolizumab in addition to gemcitabine plus cisplatin as category 1 preferred recommendations for the first-line systemic treatment of unresectable or metastatic BTCs [38].

Pembrolizumab for cHCC-CCA

Although prospective data to determine the systemic therapy for cHCC-CCA are insufficient, NCCN 2024 guideline comments that gemcitabine plus cisplatin chemotherapy combined with pembrolizumab or durvalumab is an appropriate choice for first-line therapy based on the data for HCC and CCA [38]. The subjects in the phase III KEYNOTE-966 trial that evaluated the efficacy of combination of pembrolizumab with gemcitabine and cisplatin for unresectable biliary tumors included eight cHCC-CCA patients in the pembrolizumab group and five cHCC-CCA patients in the placebo group [37]. Furthermore, case series from Korea evaluated clinical outcomes for 25 patients with advanced cHCC-CCA who were treated with pembrolizumab [39]. The median follow-up was 20.1 months (95% CI: 4.9 - 35.2 months). The median PFS was 3.5 months (95% CI: 2.4 - 4.8 months), and the median OS was 8.3 months (95% CI: 6.8 - 9.8 months). The ORR was 20.0%, meaning one out of five patients responded to pembrolizumab.

Atezolizumab and Bevacizumab▴Top 

Angiogenesis plays a key role in tumor progression process and crosstalk between angiogenesis and immune regulation in the tumor microenvironment and has gained increasing attention over the recent years [40]. Rapid tumor growth leads to hypoxia, which stimulates hypoxia-inducible factor 1 (HIF-1) and promotes vascular endothelial growth factor (VEGF)-mediated tumor angiogenesis [40, 41]. Endothelial cells of pathological vasculature have immunosuppressive features such as PD-L1 expression, which promotes T-cell exhaustion [40]. VEGF also plays immunosuppressive roles by suppressing dendric cell maturation, resulting in the interruption of T-cell priming [42], inducing exhaustion of CD8+ T cells [43], accumulating regulatory T cells (Tregs), which are the most abundant immune suppressive cells, and repolarizing tumor-associated macrophages (TAMs) to M2-like phenotypes from M1-like phenotypes [44, 45]. Meanwhile, immune cells also modulate tumor angiogenesis. Tumor-promoting cells such as Tregs enhance angiogenesis via secretion of VEGF and recruitment of endothelial cells [46]. Interestingly, Tregs express immune checkpoint molecules, such as CTLA4 and PD-1 [47]. PD-1 blockade upregulates Treg activation while CTLA4 blockage downregulates it [48]. On the other hand, tumor-suppressing cells, such as mDC, M1-like TAMs, CD8+ cells, and T helper-1 cells suppress angiogenesis [49-51]. Understanding the interactions between tumor immunity and angiogenesis has led to the exploration of combination therapy involving immunotherapy and vascular-targeting therapy. For example, preclinical studies in various cancer models showed that anti-VEGFR2 treatment upregulates the expression of PD-L1 [52, 53]. Shigeta et al reported the use of combination therapy with anti-VEGFR2 and anti-PD-1 and observed the promotion of vascular normalization and increased accumulation of tumor-suppressive CTLs and M1-like TAMs in murine HCC model [53].

Atezolizumab is a monoclonal antibody which selectively targets PD-L1, preventing interaction with receptors, PD-1 and B7-1, and reverses T-cell suppression against cancer immunity [54]. Bevacizumab is a monoclonal antibody that targets VEGF [55] and has been used for many advanced cancers in combination with chemotherapy [56, 57]. Liver is known to have a higher VEGF level compared to other organs due to hypervascularity [58], and overexpression of VEGF has been linked to the development and progression of liver cancer [59, 60]. While clinical trials combining anti-angiogenic therapy with PD-1/PD-L1 inhibitors have shown promising outcomes in non-squamous non-small cell lung cancer, renal cell carcinoma, and endometrial cancer [61-64], the combination of atezolizumab and bevacizumab has yielded the most successful results in HCC.

Atezolizumab and bevacizumab for HCC

IMbrave150 is a global, open-label, phase III trial, assessing the efficacy of combination of atezolizumab and bevacizumab for unresectable HCC as first-line treatment, which showed that the combination had better OS and PFS outcomes compared to sorafenib [65]. In this study, 501 patients at 111 sites across 17 countries were randomly assigned in a 2:1 ratio to receive either atezolizumab plus bevacizumab or sorafenib. The median follow-up time was 8.6 months. The HR for death comparing the atezolizumab plus bevacizumab group to the sorafenib group was 0.58 (95% CI: 0.42 - 0.79; P < 0.001), and OS at 12 months was 67.2% (95% CI: 61.3-73.1%) for atezolizumab plus bevacizumab and 54.6% (95% CI: 45.2-64.0%) for sorafenib. Updated analysis with a median follow-up of 15.6 months showed a median OS of 19.2 months (95% CI: 17.0 - 23.7) with atezolizumab plus bevacizumab and 13.4 months (95% CI: 11.4 - 16.9) with sorafenib (HR: 0.66; 95% CI: 0.52 - 0.85; descriptive P < 0.001) [66]. The median PFS was 6.9 months (95% CI: 5.7 - 8.6 months) for the atezolizumab plus bevacizumab group and 4.3 months (95% CI: 4.0 - 5.6 months) for the sorafenib group (HR: 0.65; 95% CI: 0.53 - 0.81; descriptive P < 0.001). Following this study, the 2024 NCCN guideline recommends the combination of atezolizumab and bevacizumab as the first-line systemic therapy for advanced HCC as a category 1 recommendation [32].

Atezolizumab and bevacizumab for cHCC-CCA

Several published studies suggest the utility of the combination of atezolizumab and bevacizumab for cHCC-CCA. A retrospective, multicentric study from France demonstrated antitumor efficacy of the combination therapy of atezolizumab and bevacizumab in patients with unresectable or metastatic cHCC-CCA [67]. In this study, 16 patients with unresectable or metastatic cHCC-CCA from seven centers were included. Nine patients received atezolizumab and bevacizumab as a first-line systemic therapy and seven patients as a second line or more. The first-line therapy group had a median OS of 13 months and a median PFS of 3 months. Among the seven patients receiving this combination therapy as a second line or more, four patients had stable disease, two had a partial response, and one had progressive disease. Saito et al reported a case of the use of atezolizumab plus bevacizumab for distant lymph node recurrence of cHCC-CCA after surgical resection [68]. The patient developed distant lymph node metastases after right hepatic lobectomy and adjuvant chemotherapy with titanium silicate (TS)-1 for cHCC-CCA. The patient initially received gemcitabine and cisplatin, but was discontinued due to severe rash. The patient then subsequently received lenvatinib, but it was also discontinued due to grade 3 adverse event, and the decision was made to switch to atezolizumab and bevacizumab. The patient achieved 7.5 months of PFS before ultimate progression of disease.

Nivolumab▴Top 

Nivolumab is a human immunoglobulin G4 monoclonal antibody targeting PD-1 and blocks the interaction between PD-1 expressed on T cells and PD-L1 expressed on tumor cells, modulating T-cell-modulated immune response [69, 70]. Nivolumab initially showed efficacy for melanoma [71, 72] and has been extensively studied for multiple cancers.

Nivolumab for HCC

CheckMate 459 is a randomized phase III trial, comparing nivolumab monotherapy to sorafenib as the frontline treatment for advanced HCC [73]. A total of 743 patients from 22 countries and territories in Asia, Australasia, Europe, and North America were randomly assigned to treatment. Median OS was 16.4 months (95% CI: 13.9 - 18.4) with nivolumab and 14.7 months (11.9 - 17.2) with sorafenib (HR: 0.85; 95% CI: 0.72 - 1.02; P = 0.075; minimum follow-up 22.8 months), but the protocol-defined significance level (P = 0.0419) was not reached in this trial. The study displayed the favorable efficiency and safety of nivolumab monotherapy, but it did not demonstrate a statistically significant OS compared to sorafenib. Although nivolumab monotherapy was removed from the recommendations as the first-line therapy for advanced HCC in version 1 of NCCN 2024 guideline, it is recommended as subsequent-line systemic therapy in certain circumstances [32]. Nivolumab has been also studied in combination with ipilimumab, a CTLA-4 ICI. Although PD-1 and CTLA-4 both inhibit T-cell activation, they act on different phases of the cancer immunity cycle. PD-1 is a major regulator of T-cell exhaustion in the tumor microenvironment while CTLA-4 mainly inhibits activated and regulatory T cells in the lymphoid organs [74, 75]. In CheckMate 040, a multicenter, open-label, multicohort, phase I/II study, efficacy of the combination therapy with nivolumab and ipilimumab was studied [76]. In this study, a total of 148 patients with advanced HCC previously treated with sorafenib from 31 centers in 10 territories in Asia, Europe, and North America were randomized across three dosing arms. The median follow-up was 30.7 months, and investigator-assessed ORR was greater than 30% across treatment arms. The arm A regimen (four doses nivolumab 1 mg/kg plus ipilimumab 3 mg/kg every 3 weeks then nivolumab 240 mg every 2 weeks) had the highest complete response rate, promising median OS of 22.8 months, 12-month survival rates of 61%, 24-month survival rates of 48%, and 30-month OS rates of 44%, receiving accelerated approval in the USA. Currently, NCCN 2024 guideline recommends the combination of nivolumab plus ipilimumab as second-line therapy for HCC [32].

Nivolumab for CCA

Nivolumab monotherapy for advanced BTC was evaluated in a multicenter, phase II trial with 54 patients [77]. In this study, patients with advanced BTCs with disease progression after treatment with at least one line but no more than three lines of systemic therapy were included. The inclusion criteria comprised patients with advanced BTCs who had experienced disease progression while undergoing treatment using at least one line but no more than three lines of systemic therapy. The study showed ORRs of 22% by investigator review and 11% by central review, and DOR of 59% by investigator review and 50% by central review per Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 criteria. In the intention-to-treat population, the median PFS and median OS were 3.7 months (95% CI: 2.3 - 5.7 months) and 14.2 months (95% CI: 6.0 months - not reached), respectively. Of note, PD-L1 expression was associated with prolonged PFS but was not significantly associated with OS (HR: 0.23; 95% CI: 0.10 - 0.51; P < 0.001). Current NCCN 2024 guideline recommends nivolumab as category 2B, useful in certain circumstances as a subsequent-line systemic therapy option for unresectable or metastatic progressive CCA without prior history of a checkpoint inhibitor use [38].

In terms of combination therapy with other ICIs, the combination of nivolumab plus ipilimumab was evaluated in the CheckMate 848 trial [78]. In this randomized, open-label, phase II clinical trial, 201 patients with advanced or metastatic solid tumors of high tumor mutational burden (TMB) were randomized 2:1 to receive the combination of nivolumab plus ipilimumab or nivolumab monotherapy. TMB is defined as the total number of somatic mutations in the tumor genome and high tissue TMB (tTMB) or high blood TMB (bTMB) tumors are thought to be more responsive to immunotherapies compared to low TMB tumors [79, 80]. Indeed, it has been shown that TMB is a predictive biomarker for response to immunotherapies in solid tumors [81, 82]. In the high tTMB cohort of this study, ORR was 38.6% (95% CI: 28.4-49.6%) with nivolumab and ipilimumab and 29.8% (95% CI: 17.3-44.9%) with nivolumab monotherapy. In the high bTMB cohort, ORR was 22.5% (95% CI: 13.9-33.2%) with nivolumab and ipilimumab and 15.6% (95% CI: 6.5-29.5%) with nivolumab monotherapy. Median OS with high tTMB was 15.0 (95% CI: 10.2 - 29.8) and 14.6 (95% CI: 7.7 - 20.7) months in the nivolumab plus ipilimumab, and nivolumab monotherapy arms, respectively; in patients with high bTMB, median OS was 8.1 (95% CI: 5.8 - 10.5) and 11.2 (95% CI: 5.3 - 19.0) months, respectively. Based on this study, NCCN 2024 recommends the combination of nivolumab and ipilimumab as first and second line for advanced BTC with high TMB [38].

Nivolumab for cHCC-CCA

NCCN 2024 guideline recommends the combination of nivolumab plus ipilimumab as a reasonable treatment based on the evidence extrapolated from treatment efficacy on both HCC and CCA [38]. Jang et al published a case series of 25 patients who received ICIs for unresectable or metastatic cHCC-CCA [39]. Seventeen patients (68%) received nivolumab. All patients, except one, had previously received systemic therapy. Median PFS was 3.5 months, median OS was 8.3 months, and ORR was 20.0%.

Durvalumab▴Top 

Durvalumab is an IgG1 monoclonal antibody that has high-affinity binding to PD-L1 receptor and was first approved by FDA for urothelial carcinoma in 2017 [83]. Durvalumab has been used alone or with combination of other ICIs including CTLA-4 inhibitors in various cancers. Of note, while the PD-1 pathway regulates T-cell activation in peripheral tissues, CTLA-4 is thought to regulate T-cell proliferation in lymph nodes [84-86]. It has been shown that CTLA-4 blockage enhances the efficacy of PD-1 inhibitors [87, 88].

Durvalumab for HCC

In the phase III HIMALAYA trial, comparison was made between tremelimumab, a CTLA-4 inhibitor, plus durvalumab, durvalumab monotherapy, and sorafenib as the first-line therapy for unresectable HCC [89]. Compared to sorafenib, the combination treatment significantly improved OS, and durvalumab monotherapy was noninferior to sorafenib. Based on the result of this study, NCCN 2024 guideline recommends the combination of tremelimumab plus durvalumab as the preferred regimen (category 1) and durvalumab monotherapy as one of the other recommended regimens (category 1) for first-line therapy [32]. This STRIDE regimen has gained approval internationally as first-line treatment for unresectable HCC, and durvalumab monotherapy has also been approved in Japan and the European Union.

Durvalumab for CCA

The TOPAZ-1 trial is a double-blind, placebo-controlled, phase III study for the addition of durvalumab to gemcitabine plus cisplatin for advanced BTC as first-line therapy [90]. A total of 685 patients including 383 (55.9%) intrahepatic CCA were randomly assigned to treatment. Median OS was 12.8 months (95% CI: 11.1 - 14.0) in the durvalumab group and 11.5 months (95% CI: 10.1 - 12.5) in the placebo group. Durvalumab significantly improved OS compared to placebo (HR: 0.80; 95% CI: 0.66 - 0.97; P = 0.021). Of note, HRs of OS and PFS for intrahepatic CCA were 0.76 (95% CI: 0.58 - 0.98) and 0.79 (95% CI: 0.64 - 0.99), respectively. NCCN 2024 guideline recommends the combination of durvalumab, gemcitabine plus cisplatin as primary treatment for unresectable and metastatic disease (category 1) [38]. This regimen is also recommended as an option for patients with recurrent disease more than 6 months after surgery or completion of adjuvant therapy.

Durvalumab for cHCC-CCA

NCCN 2024 guideline recommends the regimen of gemcitabine plus cisplatin chemotherapy with either durvalumab or pembrolizumab immunotherapy as an appropriate choice for first-line therapy [38]. Unome et al reported the use of the STRIDE regimen for unresectable cHCC-CCA based on the HIMALAYA study [89] and the TOPAZ-1 trial [90, 91]. In this study, a grade 3 maculopapular rash developed 25 days after the initiation of durvalumab and tremelimumab combination therapy, requiring steroid therapy. Rapid tumor growth was noted after two cycles.

Toripalimab▴Top 

Toripalimab is a humanized IgG4 monoclonal antibody targeting PD-1 and has been approved by FDA and China’s National Medical Products Administration (NMPA). It has shown promising efficacy and safety profiles for urologic cancer [92], melanoma [92], and gastric cancer [93]. Lenvatinib is a multi-kinase inhibitor that targets vascular endothelial growth factor receptors (VEGFRs) 1 to 3, fibroblast growth factor receptors (FGFRs) 1 to 4, platelet-derived growth factor receptor-α (PDGFRα), rearranged during transfection (RET), and stem cell factor receptor (KIT). Like many other tyrosine kinase inhibitors (TKIs), lenvatinib is known for its antitumor immunomodulatory effects. Experimental studies on mice have been shown that lenvatinib modulates antitumor immunity by decreasing TAMs and increasing cytotoxic CD8+ T cells, which are the major effectors in the antitumor immunity and enhance antitumor activity of anti-PD-1 antibody [94-96]. In fact, the combination of PD-1 inhibitor with lenvatinib is reported to be useful in several cancer types [97-100].

Toripalimab for HCC

Multiple phase II trials using the combination of TKI and toripalimab for HCC have been published. He et al reported the results of a phase II trial looking at the efficacy of the combination of lenvatinib, toripalimab plus folinic acid, fluorouracil, and oxaliplatin (FOLFOX) for HCC with extrahepatic metastasis [101]. Thirty patients were included in this study, which demonstrated 6-month PFS rate of 66.7%, with a median PFS of 9.73 months, and a median OS of 14.63 months (95% CI: 11.77 - 17.50) with ORR of 43.3%. Zhang et al reported a prospective, multicenter, phase II study assessing the efficacy of a combination of anlotinib and toripalimab as first-line therapy for unresectable HCC [102]. Thirty-one patients from two centers in China were enrolled. ORR was 29.0% by immune-related RECIST (irRECIST)/RECIST v1.1, and 32.3% by modified RECIST (mRECIST) criteria. Median PFS was 11.0 months, and median OS was 18.2 months. The most recent phase 2 trial is a multicenter trial that used the combination of toripalimab and bevacizumab as first-line treatment for advanced HCC [103]. Fifty-four patients were enrolled in this trial. ORR was 31.5% (RECIST v1.1) and 46.3% (mRECIST). Median PFS was 8.5 months (RECIST v1.1) and 9.8 months (mRECIST). The 12-month and 24-month OS rates were 77.3% and 63.5%, respectively.

Toripalimab for CCA

A phase II study of combination of toripalimab, lenvatinib, and gemcitabine and oxaliplatin (GEMOX) for advanced intrahepatic CCA as first-line therapy was published in 2023 from China [104]. Thirty patients with advanced intrahepatic CCA from a single center were enrolled. The median follow-up time was 23.5 months, and the ORR was 80%. Twenty-three patients achieved partial response, and one achieved complete response. The median OS, PFS, and DOR were 22.5, 10.2, and 11.0 months, respectively. Another study using the combination of toripalimab and a multi-targeted TKI is a phase II study of the combination of anlotinib (multi-targeted TKI) and toripalimab for advanced BTC [105]. In this study, 15 BTC patients, including nine ICC, three HCCA, and three gallbladder cancers, were enrolled. Among 15 patients, four (26.7%) received anlotinib plus toripalimab as the first-line therapy due to intolerability to chemotherapy, and 11 patients (73.3%) received the combination as second-line therapy. The first-line treatment group achieved an ORR of 50% and a disease control rate (DCR) of 100%. The second-line treatment group had an ORR of 18.2% and a DCR of 81.8%. There was no report of serious immune-related adverse events.

Toripalimab for cHCC-CCA

Xu et al reported a case of a patient who received the combination of toripalimab and lenvatinib as second-line therapy for abdominal lymph node metastasis after radical resection and adjuvant therapy with capecitabine for cHCC-CCA [106]. The patient was still undergoing treatment and PFS exceeded 27.2 months.

Conclusions▴Top 

cHCC-CCA is a rare liver cancer without standardized systemic therapy for advanced or recurrent disease. The use of ICIs, which has effect on both HCC and CCA, has been reported for cHCC-CCA, showing encouraging outcomes. Given the rareness of cHCC-CCA, prospective clinical trials for this condition may not be feasible, and one may need to rely on available data such as this narrative review.

Acknowledgments

None to declare.

Financial Disclosure

KT is supported by US NIH grant R01CA251545, R01CA250412, R01CA160688, and R37CA248018, as well as US DoD BCRP grant W81XWH-19-1-0674 and W81XWH-19-1-0111. Roswell Park Comprehensive Cancer Center is supported by NCI/NIH grant P30-CA016056.

Conflict of Interest

The authors have no potential conflict of interest to disclose.

Author Contributions

Conceptualization: K. Shichiri and K. Takabe. Writing - original draft preparation: K. Shichiri. Writing - review and editing: K. Tung, K. Takabe, and D. Bartlett. Supervision: K. Takabe and D. Bartlett. Funding acquisition: K. Takabe.

Data Availability

The authors declare that data supporting the findings of this study are available within the article.

Abbreviations

BSC: best supportive care; BTC: biliary tract cancer; bTMB: blood tumor mutational burden; CCA: cholangiocarcinoma; cHCC-CCA: combined hepatocellular-cholangiocarcinoma; CI: confidence interval; CTLA-4: cytotoxic T-lymphocyte-associated protein 4; DCR: disease control rate; DOR: duration of response; FDA: Food and Drug Administration; FGFR: fibroblast growth factor receptor; HR: hazard ratio; HCC: hepatocellular carcinoma; ICI: immune checkpoint inhibitor; MSI-H: microsatellite instability-high; MSI-H/dMMR: microsatellite instability-high/mismatch repair deficiency; MMR: mismatch repair; NCCN: National Comprehensive Cancer Network; NMPA: National Medical Products Administration; ORR: objective response rate; OS: overall survival; PD-1: programmed cell death-1; PDGFRα: platelet-derived growth factor receptor-α; PD-L1: programmed cell death-ligand 1; PD-L2: programmed cell death-ligand 2; PFS: progression-free survival; RECIST: Response Evaluation Criteria in Solid Tumors; SEER: Surveillance, Epidemiology, and End Results; TS: titanium silicate; TKI: tyrosine kinase inhibitor; TMB: tumor mutational burden; tTMB: tissue tumor mutational burden; VEGF: vascular endothelial growth factor; VEGFR: vascular endothelial growth factor receptor; WHO: World Health Organization


References▴Top 
  1. Brunt E, Aishima S, Clavien PA, Fowler K, Goodman Z, Gores G, Gouw A, et al. cHCC-CCA: Consensus terminology for primary liver carcinomas with both hepatocytic and cholangiocytic differentation. Hepatology. 2018;68(1):113-126.
    doi pubmed
  2. Board WCoTE. Lyon: International Agency for Research on Cancer; 2019.
  3. Ramai D, Ofosu A, Lai JK, Reddy M, Adler DG. Combined hepatocellular cholangiocarcinoma: a population-based retrospective study. Am J Gastroenterol. 2019;114(9):1496-1501.
    doi pubmed
  4. Garancini M, Goffredo P, Pagni F, Romano F, Roman S, Sosa JA, Giardini V. Combined hepatocellular-cholangiocarcinoma: a population-level analysis of an uncommon primary liver tumor. Liver Transpl. 2014;20(8):952-959.
    doi pubmed
  5. Jarnagin WR, Weber S, Tickoo SK, Koea JB, Obiekwe S, Fong Y, DeMatteo RP, et al. Combined hepatocellular and cholangiocarcinoma: demographic, clinical, and prognostic factors. Cancer. 2002;94(7):2040-2046.
    doi pubmed
  6. Lee CC, Wu CY, Chen JT, Chen GH. Comparing combined hepatocellular-cholangiocarcinoma and cholangiocarcinoma: a clinicopathological study. Hepatogastroenterology. 2002;49(48):1487-1490.
    pubmed
  7. Wells HG. Primary carcinoma of liver. The American Journal of the Medical Sciences. 1903;126(3):403-417.
  8. Yano Y, Yamamoto J, Kosuge T, Sakamoto Y, Yamasaki S, Shimada K, Ojima H, et al. Combined hepatocellular and cholangiocarcinoma: a clinicopathologic study of 26 resected cases. Jpn J Clin Oncol. 2003;33(6):283-287.
    doi pubmed
  9. Yin X, Zhang BH, Qiu SJ, Ren ZG, Zhou J, Chen XH, Zhou Y, et al. Combined hepatocellular carcinoma and cholangiocarcinoma: clinical features, treatment modalities, and prognosis. Ann Surg Oncol. 2012;19(9):2869-2876.
    doi pubmed
  10. Zhang G, Chen BW, Yang XB, Wang HY, Yang X, Xie FC, Chen XQ, et al. Prognostic analysis of patients with combined hepatocellular-cholangiocarcinoma after radical resection: A retrospective multicenter cohort study. World J Gastroenterol. 2022;28(41):5968-5981.
    doi pubmed
  11. Tang Q, Chen Y, Li X, Long S, Shi Y, Yu Y, Wu W, et al. The role of PD-1/PD-L1 and application of immune-checkpoint inhibitors in human cancers. Front Immunol. 2022;13:964442.
    doi pubmed
  12. Tokumaru Y, Joyce D, Takabe K. Current status and limitations of immunotherapy for breast cancer. Surgery. 2020;167(3):628-630.
    doi pubmed
  13. Gupta RK, Roy AM, Gupta A, Takabe K, Dhakal A, Opyrchal M, Kalinski P, et al. Systemic therapy de-escalation in early-stage triple-negative breast cancer: dawn of a new era? Cancers (Basel). 2022;14(8):1856.
    doi pubmed
  14. Vogel A, Meyer T, Sapisochin G, Salem R, Saborowski A. Hepatocellular carcinoma. Lancet. 2022;400(10360):1345-1362.
    doi pubmed
  15. Llovet JM, Castet F, Heikenwalder M, Maini MK, Mazzaferro V, Pinato DJ, Pikarsky E, et al. Immunotherapies for hepatocellular carcinoma. Nat Rev Clin Oncol. 2022;19(3):151-172.
    doi pubmed
  16. Byun DJ, Wolchok JD, Rosenberg LM, Girotra M. Cancer immunotherapy - immune checkpoint blockade and associated endocrinopathies. Nat Rev Endocrinol. 2017;13(4):195-207.
    doi pubmed
  17. Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, Lu S, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357(6349):409-413.
    doi pubmed
  18. Marabelle A, Le DT, Ascierto PA, Di Giacomo AM, De Jesus-Acosta A, Delord JP, Geva R, et al. Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair-deficient cancer: results from the phase II KEYNOTE-158 study. J Clin Oncol. 2020;38(1):1-10.
    doi pubmed
  19. Yu I, Dakwar A, Takabe K. Immunotherapy: recent advances and its future as a neoadjuvant, adjuvant, and primary treatment in colorectal cancer. Cells. 2023;12(2).
    doi pubmed
  20. Scapin G, Yang X, Prosise WW, McCoy M, Reichert P, Johnston JM, Kashi RS, et al. Structure of full-length human anti-PD1 therapeutic IgG4 antibody pembrolizumab. Nat Struct Mol Biol. 2015;22(12):953-958.
    doi pubmed
  21. Blank C, Brown I, Peterson AC, Spiotto M, Iwai Y, Honjo T, Gajewski TF. PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell receptor (TCR) transgenic CD8+ T cells. Cancer Res. 2004;64(3):1140-1145.
    doi pubmed
  22. Chuk MK, Chang JT, Theoret MR, Sampene E, He K, Weis SL, Helms WS, et al. FDA approval summary: accelerated approval of pembrolizumab for second-line treatment of metastatic melanoma. Clin Cancer Res. 2017;23(19):5666-5670.
    doi pubmed
  23. Marcus L, Lemery SJ, Keegan P, Pazdur R. FDA approval summary: pembrolizumab for the treatment of microsatellite instability-high solid tumors. Clin Cancer Res. 2019;25(13):3753-3758.
    doi pubmed
  24. Pai-Scherf L, Blumenthal GM, Li H, Subramaniam S, Mishra-Kalyani PS, He K, Zhao H, et al. FDA approval summary: pembrolizumab for treatment of metastatic non-small cell lung cancer: first-line therapy and beyond. Oncologist. 2017;22(11):1392-1399.
    doi pubmed
  25. Larkins E, Blumenthal GM, Yuan W, He K, Sridhara R, Subramaniam S, Zhao H, et al. FDA approval summary: pembrolizumab for the treatment of recurrent or metastatic head and neck squamous cell carcinoma with disease progression on or after platinum-containing chemotherapy. Oncologist. 2017;22(7):873-878.
    doi pubmed
  26. Fashoyin-Aje L, Donoghue M, Chen H, He K, Veeraraghavan J, Goldberg KB, Keegan P, et al. FDA approval summary: pembrolizumab for recurrent locally advanced or metastatic gastric or gastroesophageal junction adenocarcinoma expressing PD-L1. Oncologist. 2019;24(1):103-109.
    doi pubmed
  27. Suzman DL, Agrawal S, Ning YM, Maher VE, Fernandes LL, Karuri S, Tang S, et al. FDA approval summary: atezolizumab or pembrolizumab for the treatment of patients with advanced urothelial carcinoma ineligible for cisplatin-containing chemotherapy. Oncologist. 2019;24(4):563-569.
    doi pubmed
  28. Zhu AX, Finn RS, Edeline J, Cattan S, Ogasawara S, Palmer D, Verslype C, et al. Pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib (KEYNOTE-224): a non-randomised, open-label phase 2 trial. Lancet Oncol. 2018;19(7):940-952.
    doi pubmed
  29. Finn RS, Ryoo BY, Merle P, Kudo M, Bouattour M, Lim HY, Breder V, et al. Pembrolizumab as second-line therapy in patients with advanced hepatocellular carcinoma in KEYNOTE-240: a randomized, double-blind, phase III trial. J Clin Oncol. 2020;38(3):193-202.
    doi pubmed
  30. Qin S, Chen Z, Fang W, Ren Z, Xu R, Ryoo BY, Meng Z, et al. Pembrolizumab versus placebo as second-line therapy in patients from asia with advanced hepatocellular carcinoma: a randomized, double-blind, phase III trial. J Clin Oncol. 2023;41(7):1434-1443.
    doi pubmed
  31. Verset G, Borbath I, Karwal M, Verslype C, Van Vlierberghe H, Kardosh A, Zagonel V, et al. Pembrolizumab monotherapy for previously untreated advanced hepatocellular carcinoma: data from the open-label, phase II KEYNOTE-224 trial. Clin Cancer Res. 2022;28(12):2547-2554.
    doi pubmed
  32. Network NCC. Clinical practice guidelines in oncology: hepatocellular carcinoma. Version 2.2024. 2024.
  33. Maio M, Ascierto PA, Manzyuk L, Motola-Kuba D, Penel N, Cassier PA, Bariani GM, et al. Pembrolizumab in microsatellite instability high or mismatch repair deficient cancers: updated analysis from the phase II KEYNOTE-158 study. Ann Oncol. 2022;33(9):929-938.
    doi pubmed
  34. Zitvogel L, Galluzzi L, Smyth MJ, Kroemer G. Mechanism of action of conventional and targeted anticancer therapies: reinstating immunosurveillance. Immunity. 2013;39(1):74-88.
    doi pubmed
  35. de Biasi AR, Villena-Vargas J, Adusumilli PS. Cisplatin-induced antitumor immunomodulation: a review of preclinical and clinical evidence. Clin Cancer Res. 2014;20(21):5384-5391.
    doi pubmed
  36. Li XM, Zhang XM, Li JY, Jiang N, Chen L, Tang LL, Mao YP, et al. The immune modulation effects of gemcitabine plus cisplatin induction chemotherapy in nasopharyngeal carcinoma. Cancer Med. 2022;11(18):3437-3444.
    doi pubmed
  37. Kelley RK, Ueno M, Yoo C, Finn RS, Furuse J, Ren Z, Yau T, et al. Pembrolizumab in combination with gemcitabine and cisplatin compared with gemcitabine and cisplatin alone for patients with advanced biliary tract cancer (KEYNOTE-966): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2023;401(10391):1853-1865.
    doi pubmed
  38. Network NCC. Clinical practice guidelines in oncology: biliary tract cancers, Version 2.2024. 2024.
  39. Jang YJ, Kim EJ, Kim HD, Kim KP, Ryu MH, Park SR, Choi WM, et al. Clinical outcomes of immune checkpoint inhibitors in unresectable or metastatic combined hepatocellular-cholangiocarcinoma. J Cancer Res Clin Oncol. 2023;149(10):7547-7555.
    doi pubmed
  40. Lee WS, Yang H, Chon HJ, Kim C. Combination of anti-angiogenic therapy and immune checkpoint blockade normalizes vascular-immune crosstalk to potentiate cancer immunity. Exp Mol Med. 2020;52(9):1475-1485.
    doi pubmed
  41. Jain RK. Antiangiogenesis strategies revisited: from starving tumors to alleviating hypoxia. Cancer Cell. 2014;26(5):605-622.
    doi pubmed
  42. Gabrilovich DI, Chen HL, Girgis KR, Cunningham HT, Meny GM, Nadaf S, Kavanaugh D, et al. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat Med. 1996;2(10):1096-1103.
    doi pubmed
  43. Kim CG, Jang M, Kim Y, Leem G, Kim KH, Lee H, Kim TS, et al. VEGF-A drives TOX-dependent T cell exhaustion in anti-PD-1-resistant microsatellite stable colorectal cancers. Sci Immunol. 2019;4(41):eaay0555.
    doi pubmed
  44. Fukumura D, Kloepper J, Amoozgar Z, Duda DG, Jain RK. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat Rev Clin Oncol. 2018;15(5):325-340.
    doi pubmed
  45. Huang Y, Kim BYS, Chan CK, Hahn SM, Weissman IL, Jiang W. Improving immune-vascular crosstalk for cancer immunotherapy. Nat Rev Immunol. 2018;18(3):195-203.
    doi pubmed
  46. DeNardo DG, Barreto JB, Andreu P, Vasquez L, Tawfik D, Kolhatkar N, Coussens LM. CD4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell. 2009;16(2):91-102.
    doi pubmed
  47. Granito A, Muratori L, Lalanne C, Quarneti C, Ferri S, Guidi M, Lenzi M, et al. Hepatocellular carcinoma in viral and autoimmune liver diseases: Role of CD4+ CD25+ Foxp3+ regulatory T cells in the immune microenvironment. World J Gastroenterol. 2021;27(22):2994-3009.
    doi pubmed
  48. Tay C, Tanaka A, Sakaguchi S. Tumor-infiltrating regulatory T cells as targets of cancer immunotherapy. Cancer Cell. 2023;41(3):450-465.
    doi pubmed
  49. Qian BZ, Pollard JW. Macrophage diversity enhances tumor progression and metastasis. Cell. 2010;141(1):39-51.
    doi pubmed
  50. Tian L, Goldstein A, Wang H, Ching Lo H, Sun Kim I, Welte T, Sheng K, et al. Mutual regulation of tumour vessel normalization and immunostimulatory reprogramming. Nature. 2017;544(7649):250-254.
    doi pubmed
  51. Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat Rev Immunol. 2003;3(2):133-146.
    doi pubmed
  52. Allen E, Jabouille A, Rivera LB, Lodewijckx I, Missiaen R, Steri V, Feyen K, et al. Combined antiangiogenic and anti-PD-L1 therapy stimulates tumor immunity through HEV formation. Sci Transl Med. 2017;9(385):eaak9679.
    doi pubmed
  53. Shigeta K, Datta M, Hato T, Kitahara S, Chen IX, Matsui A, Kikuchi H, et al. Dual programmed death receptor-1 and vascular endothelial growth factor receptor-2 blockade promotes vascular normalization and enhances antitumor immune responses in hepatocellular carcinoma. Hepatology. 2020;71(4):1247-1261.
    doi pubmed
  54. Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, Sosman JA, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515(7528):563-567.
    doi pubmed
  55. Ferrara N, Hillan KJ, Novotny W. Bevacizumab (Avastin), a humanized anti-VEGF monoclonal antibody for cancer therapy. Biochem Biophys Res Commun. 2005;333(2):328-335.
    doi pubmed
  56. Garcia J, Hurwitz HI, Sandler AB, Miles D, Coleman RL, Deurloo R, Chinot OL. Bevacizumab (Avastin(R)) in cancer treatment: A review of 15 years of clinical experience and future outlook. Cancer Treat Rev. 2020;86:102017.
    doi pubmed
  57. Stevenson CE, Nagahashi M, Ramachandran S, Yamada A, Bear HD, Takabe K. Bevacizumab and breast cancer: what does the future hold? Future Oncol. 2012;8(4):403-414.
    doi pubmed
  58. Brodsky SV, Mendelev N, Melamed M, Ramaswamy G. Vascular density and VEGF expression in hepatic lesions. J Gastrointestin Liver Dis. 2007;16(4):373-377.
    pubmed
  59. Zhu AX, Duda DG, Sahani DV, Jain RK. HCC and angiogenesis: possible targets and future directions. Nat Rev Clin Oncol. 2011;8(5):292-301.
    doi pubmed
  60. Morse MA, Sun W, Kim R, He AR, Abada PB, Mynderse M, Finn RS. The role of angiogenesis in hepatocellular carcinoma. Clin Cancer Res. 2019;25(3):912-920.
    doi pubmed
  61. Makker V, Rasco D, Vogelzang NJ, Brose MS, Cohn AL, Mier J, Di Simone C, et al. Lenvatinib plus pembrolizumab in patients with advanced endometrial cancer: an interim analysis of a multicentre, open-label, single-arm, phase 2 trial. Lancet Oncol. 2019;20(5):711-718.
    doi pubmed
  62. Motzer RJ, Penkov K, Haanen J, Rini B, Albiges L, Campbell MT, Venugopal B, et al. Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019;380(12):1103-1115.
    doi pubmed
  63. Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, Pouliot F, et al. Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019;380(12):1116-1127.
    doi pubmed
  64. Socinski MA, Jotte RM, Cappuzzo F, Orlandi F, Stroyakovskiy D, Nogami N, Rodriguez-Abreu D, et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N Engl J Med. 2018;378(24):2288-2301.
    doi pubmed
  65. Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, Kudo M, et al. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med. 2020;382(20):1894-1905.
    doi pubmed
  66. Cheng AL, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, Lim HY, et al. Updated efficacy and safety data from IMbrave150: Atezolizumab plus bevacizumab vs. sorafenib for unresectable hepatocellular carcinoma. J Hepatol. 2022;76(4):862-873.
    doi pubmed
  67. Gigante E, Bouattour M, Bedoya JU, Regnault H, Ziol M, Assenat E, Paradis V, et al. Atezolizumab and bevacizumab for non-resectable or metastatic combined hepatocellular-cholangiocarcinoma: A multicentric retrospective study. United European Gastroenterol J. 2024;12(4):429-439.
    doi pubmed
  68. Saito N, Hatanaka T, Nakano S, Hazama Y, Yoshida S, Hachisu Y, Tanaka Y, et al. A case of unresectable combined hepatocellular and cholangiocarcinoma treated with atezolizumab plus bevacizumab. Clin Case Rep. 2022;10(7):e6129.
    doi pubmed
  69. Wang C, Thudium KB, Han M, Wang XT, Huang H, Feingersh D, Garcia C, et al. In vitro characterization of the anti-PD-1 antibody nivolumab, BMS-936558, and in vivo toxicology in non-human primates. Cancer Immunol Res. 2014;2(9):846-856.
    doi pubmed
  70. Chen L, Han X. Anti-PD-1/PD-L1 therapy of human cancer: past, present, and future. J Clin Invest. 2015;125(9):3384-3391.
    doi pubmed
  71. Weber JS, D'Angelo SP, Minor D, Hodi FS, Gutzmer R, Neyns B, Hoeller C, et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2015;16(4):375-384.
    doi pubmed
  72. Larkin J, Minor D, D'Angelo S, Neyns B, Smylie M, Miller WH, Jr., Gutzmer R, et al. Overall survival in patients with advanced melanoma who received nivolumab versus investigator's choice chemotherapy in CheckMate 037: a randomized, controlled, open-label phase iii trial. J Clin Oncol. 2018;36(4):383-390.
    doi pubmed
  73. Yau T, Park JW, Finn RS, Cheng AL, Mathurin P, Edeline J, Kudo M, et al. Nivolumab versus sorafenib in advanced hepatocellular carcinoma (CheckMate 459): a randomised, multicentre, open-label, phase 3 trial. Lancet Oncol. 2022;23(1):77-90.
    doi pubmed
  74. Das R, Verma R, Sznol M, Boddupalli CS, Gettinger SN, Kluger H, Callahan M, et al. Combination therapy with anti-CTLA-4 and anti-PD-1 leads to distinct immunologic changes in vivo. J Immunol. 2015;194(3):950-959.
    doi pubmed
  75. Melero I, Berman DM, Aznar MA, Korman AJ, Perez Gracia JL, Haanen J. Evolving synergistic combinations of targeted immunotherapies to combat cancer. Nat Rev Cancer. 2015;15(8):457-472.
    doi pubmed
  76. Yau T, Kang YK, Kim TY, El-Khoueiry AB, Santoro A, Sangro B, Melero I, et al. Efficacy and safety of nivolumab plus ipilimumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib: the CheckMate 040 randomized clinical trial. JAMA Oncol. 2020;6(11):e204564.
    doi pubmed
  77. Kim RD, Chung V, Alese OB, El-Rayes BF, Li D, Al-Toubah TE, Schell MJ, et al. A phase 2 multi-institutional study of nivolumab for patients with advanced refractory biliary tract cancer. JAMA Oncol. 2020;6(6):888-894.
    doi pubmed
  78. Schenker M, Burotto M, Richardet M, Ciuleanu TE, Goncalves A, Steeghs N, Schoffski P, et al. Randomized, open-label, phase 2 study of nivolumab plus ipilimumab or nivolumab monotherapy in patients with advanced or metastatic solid tumors of high tumor mutational burden. J Immunother Cancer. 2024;12(8):e008872.
    doi pubmed
  79. Cao J, Yang X, Chen S, Wang J, Fan X, Fu S, Yang L. The predictive efficacy of tumor mutation burden in immunotherapy across multiple cancer types: a meta-analysis and bioinformatics analysis. Transl Oncol. 2022;20:101375.
    doi pubmed
  80. Budczies J, Kazdal D, Menzel M, Beck S, Kluck K, Altburger C, Schwab C, et al. Tumour mutational burden: clinical utility, challenges and emerging improvements. Nat Rev Clin Oncol. 2024;21(10):725-742.
    doi pubmed
  81. Marabelle A, Fakih M, Lopez J, Shah M, Shapira-Frommer R, Nakagawa K, Chung HC, et al. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol. 2020;21(10):1353-1365.
    doi pubmed
  82. Hellmann MD, Callahan MK, Awad MM, Calvo E, Ascierto PA, Atmaca A, Rizvi NA, et al. Tumor mutational burden and efficacy of nivolumab monotherapy and in combination with ipilimumab in small-cell lung cancer. Cancer Cell. 2019;35(2):329.
    doi pubmed
  83. Syed YY. Erratum to: Durvalumab: First Global Approval. Drugs. 2017;77(16):1817.
    doi pubmed
  84. Fife BT, Bluestone JA. Control of peripheral T-cell tolerance and autoimmunity via the CTLA-4 and PD-1 pathways. Immunol Rev. 2008;224:166-182.
    doi pubmed
  85. Krummel MF, Allison JP. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J Exp Med. 1995;182(2):459-465.
    doi pubmed
  86. Buchbinder EI, Desai A. CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition. Am J Clin Oncol. 2016;39(1):98-106.
    doi pubmed
  87. Zhao Y, Lee CK, Lin CH, Gassen RB, Xu X, Huang Z, Xiao C, et al. PD-L1:CD80 cis-heterodimer triggers the co-stimulatory receptor CD28 while repressing the inhibitory PD-1 and CTLA-4 pathways. Immunity. 2019;51(6):1059-1073.e1059.
    doi pubmed
  88. Kudo M. Scientific rationale for combination immunotherapy of hepatocellular carcinoma with anti-PD-1/PD-L1 and anti-CTLA-4 antibodies. Liver Cancer. 2019;8(6):413-426.
    doi pubmed
  89. Sangro B, Chan SL, Kelley RK, Lau G, Kudo M, Sukeepaisarnjaroen W, Yarchoan M, et al. Four-year overall survival update from the phase III HIMALAYA study of tremelimumab plus durvalumab in unresectable hepatocellular carcinoma. Ann Oncol. 2024;35(5):448-457.
    doi pubmed
  90. Oh DY, Ruth He A, Qin S, Chen LT, Okusaka T, Vogel A, Kim JW, et al. Durvalumab plus gemcitabine and cisplatin in advanced biliary tract cancer. NEJM Evid. 2022;1(8):EVIDoa2200015.
    doi pubmed
  91. Unome S, Imai K, Miwa T, Hanai T, Suetsugu A, Takai K, Suzui N, et al. Unresectable Combined Hepatocellular-cholangiocarcinoma Treated with Combination Therapy Consisting of Durvalumab Plus Tremelimumab. Intern Med. 2024;63(19):2631-2636.
    doi pubmed
  92. Tang B, Yan X, Sheng X, Si L, Cui C, Kong Y, Mao L, et al. Safety and clinical activity with an anti-PD-1 antibody JS001 in advanced melanoma or urologic cancer patients. J Hematol Oncol. 2019;12(1):7.
    doi pubmed
  93. Wang F, Wei XL, Wang FH, Xu N, Shen L, Dai GH, Yuan XL, et al. Safety, efficacy and tumor mutational burden as a biomarker of overall survival benefit in chemo-refractory gastric cancer treated with toripalimab, a PD-1 antibody in phase Ib/II clinical trial NCT02915432. Ann Oncol. 2019;30(9):1479-1486.
    doi pubmed
  94. Kato Y, Tabata K, Kimura T, Yachie-Kinoshita A, Ozawa Y, Yamada K, Ito J, et al. Lenvatinib plus anti-PD-1 antibody combination treatment activates CD8+ T cells through reduction of tumor-associated macrophage and activation of the interferon pathway. PLoS One. 2019;14(2):e0212513.
    doi pubmed
  95. Kimura T, Kato Y, Ozawa Y, Kodama K, Ito J, Ichikawa K, Yamada K, et al. Immunomodulatory activity of lenvatinib contributes to antitumor activity in the Hepa1-6 hepatocellular carcinoma model. Cancer Sci. 2018;109(12):3993-4002.
    doi pubmed
  96. Stefanini B, Ielasi L, Chen R, Abbati C, Tonnini M, Tovoli F, Granito A. TKIs in combination with immunotherapy for hepatocellular carcinoma. Expert Rev Anticancer Ther. 2023;23(3):279-291.
    doi pubmed
  97. Taylor MH, Lee CH, Makker V, Rasco D, Dutcus CE, Wu J, Stepan DE, et al. Phase IB/II trial of lenvatinib plus pembrolizumab in patients with advanced renal cell carcinoma, endometrial cancer, and other selected advanced solid tumors. J Clin Oncol. 2020;38(11):1154-1163.
    doi pubmed
  98. Makker V, Colombo N, Casado Herraez A, Santin AD, Colomba E, Miller DS, Fujiwara K, et al. Lenvatinib plus pembrolizumab for advanced endometrial cancer. N Engl J Med. 2022;386(5):437-448.
    doi pubmed
  99. Makker V, Colombo N, Casado Herraez A, Monk BJ, Mackay H, Santin AD, Miller DS, et al. Lenvatinib plus pembrolizumab in previously treated advanced endometrial cancer: updated efficacy and safety from the randomized phase III study 309/KEYNOTE-775. J Clin Oncol. 2023;41(16):2904-2910.
    doi pubmed
  100. Choueiri TK, Eto M, Motzer R, De Giorgi U, Buchler T, Basappa NS, Mendez-Vidal MJ, et al. Lenvatinib plus pembrolizumab versus sunitinib as first-line treatment of patients with advanced renal cell carcinoma (CLEAR): extended follow-up from the phase 3, randomised, open-label study. Lancet Oncol. 2023;24(3):228-238.
    doi pubmed
  101. He M, Huang Y, Du Z, Lai Z, Ouyang H, Shen J, Wen D, et al. Lenvatinib, toripalimab plus FOLFOX chemotherapy in hepatocellular carcinoma patients with extrahepatic metastasis: a biomolecular exploratory, phase II trial (LTSC). Clin Cancer Res. 2023;29(24):5104-5115.
    doi pubmed
  102. Zhang CS, Zeng ZM, Zhuo MY, Luo JR, Zhuang XH, Xu JN, Zeng J, et al. Anlotinib combined with toripalimab as first-line therapy for unresectable hepatocellular carcinoma: a prospective, multicenter, phase II study. Oncologist. 2023;28(12):e1239-e1247.
    doi pubmed
  103. Chen Y, Du C, Shen S, Zhang W, Shan Y, Lyu A, Wu J, et al. Toripalimab plus bevacizumab as first-line treatment for advanced hepatocellular carcinoma: a prospective, multicenter, single-arm, phase II trial. Clin Cancer Res. 2024;30(14):2937-2944.
    doi pubmed
  104. Shi GM, Huang XY, Wu D, Sun HC, Liang F, Ji Y, Chen Y, et al. Toripalimab combined with lenvatinib and GEMOX is a promising regimen as first-line treatment for advanced intrahepatic cholangiocarcinoma: a single-center, single-arm, phase 2 study. Signal Transduct Target Ther. 2023;8(1):106.
    doi pubmed
  105. Zhou M, Jin Y, Zhu S, Xu C, Li L, Liu B, Shen J. A phase II study to evaluate the safety and efficacy of anlotinib combined with toripalimab for advanced biliary tract cancer. Clin Transl Immunology. 2024;13(1):e1483.
    doi pubmed
  106. Xu Z, Ma J, Chen T, Yang Y. Remarkable response of PD-1 antibody plus lenvatinib as the second-line treatment for combined hepatocellular-cholangiocarcinoma: Report of two cases. Asian J Surg. 2023;46(4):1741-1742.
    doi pubmed


This article is distributed under the terms of the Creative Commons Attribution Non-Commercial 4.0 International License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.


World Journal of Oncology is published by Elmer Press Inc.