Development and challenges in the treatment of advanced gallbladder cancer (Review)

  • Authors:
    • Bo Zhou
    • Shi-Ran Zhang
    • Geng Chen
  • View Affiliations

  • Published online on: June 3, 2025     https://doi.org/10.3892/ol.2025.15128
  • Article Number: 382
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gallbladder cancer (GBC) is a highly lethal disease and the most common cancer of the biliary tract. Numerous patients are diagnosed at an advanced stage, limiting surgical options and necessitating local or systemic treatment. Despite advancements in cancer treatment, 5‑year survival rates remain low. Various therapeutic methods are available for the treatment of advanced GBC, including local therapy, chemoradiotherapy, immunotherapy and targeted therapy. Although these treatments offer hope and broader prospects for patients, challenges persist. Through a systematic review of the literature from PubMed and Web of Science over the past decade, a summary and analysis of treatment trends, existing challenges and the latest research developments in advanced GBC were provided.

Introduction

Gallbladder cancer (GBC) is a highly lethal disease and the most common malignancy of the biliary tract (13). The highest incidence rates are observed in Chile and India, followed by Eastern Europe, Pakistan and Japan (4). In the United States, >1,000 new cases of GBC are diagnosed annually, with a mean age at diagnosis of 71 years (5). Women account for ~70% of GBC cases, with a female-to-male ratio of 2.41:1 (6). GBC is characterized by aggressive behavior, including local invasion, early lymph node metastasis and frequent distant metastasis. The initial stages of GBC are asymptomatic, leading to diagnoses at advanced stages (1,7,8). The median survival time is <1 year and the overall survival (OS) rate ranges from 17.8 to 21.7% (9). Although notable advancements have been made in the treatment of advanced GBC in recent years, their impact on patient prognosis remains limited. The present study aims to provide a comprehensive review of the latest treatment strategies and research progress in the management of advanced GBC.

Local treatment

Local therapies have proven effectiveness in prolonging patient survival and enhancing quality of life in advanced GBC. Transarterial hepatic embolization, transarterial chemoembolization (TACE), drug-eluting bead TACE (DEB-TACE) and hepatic arterial infusion chemotherapy (HAIC) have been well documented in the treatment of advanced GBC (1012). These treatments selectively block the tumor arterial blood supply using various embolic agents, with or without local chemotherapeutic agents (13). In the literature, the most common adverse events of vascular interventional therapy included post-embolization syndrome, characterized by abdominal pain, fever and intestinal obstruction (14). Additionally, some cases of GBC were poorly responsive to treatment. A meta-analysis of different transarterial chemotherapy methods, such as TACE, HAIC and DEB-TACE, demonstrated an OS time of 15.7 months from the time of diagnosis. These findings suggest that transarterial chemotherapy-based treatment for cholangiocarcinoma (including GBC) is safe, well-tolerated and results in a survival benefit of 2–7 months compared with systemic treatments (15).

Other local treatment options for GBC include endoscopic radiofrequency ablation (RFA), radiotherapy (RT) and endoscopic photodynamic therapy (PDT) (16,17). RFA is increasingly being utilized for local tumor control (18). Biliary tract cancers (BTCs) (including GBC) may need higher doses in order to achieve better local control and maybe also a survival benefit, compared to other solid tumors. Concepts for safer dose escalation include the use of stereotactic body RT (SBRT), brachytherapy or proton beam RT (1921). SBRT has been reported as a promising local treatment for cancer, providing effective local control (LC) and prolonging OS time. However, current guidelines recommend TACE as a primary treatment option for patients with early to intermediate-stage cancer, while SBRT is not included as a standard treatment (22). Compared with TACE, SBRT has demonstrated comparable OS time and improved LC across various stages of cancer, ranging from early to advanced stages, without causing serious adverse events. SBRT should be considered an effective treatment option for different stages of gallbladder cancer, as well as a comprehensive assessment of the patient's body. In one study, SBRT led to local control rates ranging from 65 to 100%, with OS time of 11–35.5 months (median, 15 months), in selected patients (23). RT is widely used to treat various cancer types, and while it can effectively control the local spread of GBC, it may not be sufficient on its own. To overcome the limitations of single-modality RT, combination chemotherapy is often administered concurrently.

PDT involves the intravenous injection of a photosensitive agent followed by irradiation with a specific wavelength of light to selectively target cancer cells and modulate the cancer microenvironment (24). However, the efficacy and safety of PDT for advanced GBC have not been fully evaluated. Further research is necessary to assess the role of PDT in combination with other therapies and its potential impact on the tumor microenvironment.

While there are various local treatment options for advanced GBC, each center may have different experiences and preferences with regard to selecting the most appropriate approach. Currently, there is no unified consensus on the optimal local treatment methods for advanced GBC.

Chemotherapy and radioembolization

GBC is known for its high rate of malignancy and multidrug resistance. Common chemical drugs used for the treatment of GBC include cisplatin (CDDP), gemcitabine (GEM), 5-fluorouracil (5-FU), epirubicin, oxaliplatin (OXA), S-1, capecitabine and irinotecan (2528). The results of clinical trials showed that the response rates from combining these chemotherapy drug combinations were still low at 26.1, 15.5, 30.7, 14.3, and 29.8 for GEM + CDDP, GEM, GEM + OXA, 5-FU + folinic acid and GEM + S-1, respectively (2931). The response rate of GEM + CDDP as a first-line chemotherapy regimen (26.1%) was 10.6% higher than that of GEM alone (15.5%) (29). Chemotherapy response remains suboptimal in patients with advanced GBC, often accompanied by high rates of chemotherapy resistance. Genetic testing of individual cancers that will be matched with known chemotherapy regimens or sensitizers will allow for individualized treatment based on the characteristics of specific tumor genes (32). A comprehensive list of clinical trials focusing on chemotherapy for patients with GBC was obtained from ClinicalTrials.gov and is detailed in Table I.

Table I.

Clinical trials of chemotherapy including GBC obtained from ClinicalTrials.gov.

Table I.

Clinical trials of chemotherapy including GBC obtained from ClinicalTrials.gov.

NCT numberYearStudy phaseConditionStudy size, nTreatment agentsPrimary end pointStatus
NCT028678652016-2027II/IIIGBC314RT + Gem + Cis vs. Gem + CisOSRecruitment
NCT034735742018-2022IIBTC (GBC)128Durvalumab + tremelimumab + Gem/Gem + Cis vs. Gem + CisORRNone
NCT043624492019-2023IIBTC (GBC)30Gem + CisOSRecruitment
NCT059190952023-2025IIGBC37Carrilizumab + GEMOXOS, PFSRecruitment
NCT054939562019-2023IIIGBC120Gem + Cis vs. Gem + Cis + CTRTOSRecruitment
NCT037684142019-2024IIIBTC (GBC)452Gem + Cis + nab-paclitaxel vs. Gem + CisOSRecruitment
NCT048567612020-2024IIBTC (GBC)160Capecitabine + S-1PFSRecruitment
NCT014704432011-2020IIIBTC (GBC)240GEMOX vs. XELOXPFSNone
NCT055069432023-2024II/IIIBTC (GBC)150CTX-009 + paclitaxel vs. paclitaxelORRRecruitment

[i] NCT, National Clinical Trial; BTC, biliary tract cancer; Cis, cisplatin; GBC, gallbladder cancer; Gem, gemcitabine; GEMOX, gemcitabine plus oxaliplatin; XELOX, capecitabine plus oxaliplatin; OS, overall survival; ORR, overall response rate; PFS, progression-free survival; RT, radiotherapy.

The GEM + CDDP regimen has served as the standard first-line treatment for GBC for over a decade, as demonstrated in the phase III clinical trial known as ABC-02 (29). However, owing to the risk of renal dysfunction associated with CDDP, CDDP has been gradually replaced by OXA. A single-arm phase II trial (SWOG S0809), conducted in the United States and published in 2015, enrolled 79 patients with extrahepatic cholangiocarcinoma and GBC. The trial demonstrated the efficacy of adjuvant GEM + capecitabine chemotherapy, followed by capecitabine + RT. The results showed that the 2-year survival rate was 65% (95% CI, 53–74%), with mortality rates of 67 and 60% for R0 and R1 patients, respectively. The median OS time was 35 months (R0, 34 months; R1, 35 months) (33).

Second-line or subsequent systemic therapy regimens for GBC are similar to those employed in other gastrointestinal malignancies, including regimens such as 5-FU + leucovorin + OXA (also known as FOLFOX), 5-FU + leucovorin + irinotecan (also known as FOLFIRI) and nanoliposomal irinotecan + 5-FU + leucovorin (1,34). The 2015 prospective case series reported by Dodagoudar et al (35) utilized second-line agents, including folic acid, 5-FU and oxaliplatin (FOLFOX), demonstrating promising responses. The response rate was 24.2% (16/66), the disease control rate was 59.1% (39/66), the median time to progression was 3.9 months (95% CI, 3.1–4.7) and the median OS was 7.6 months (95% CI, 6.8–8.2). RT has proven effective for patients with advanced GBC. A Phase 2 clinical trial involving 41 patients with intrahepatic cholangiocarcinoma with selective radiotherapy combined with chemotherapy (cisplatin and gemcitabine) showed a 39% increase in response rate, a median progression-free survival (PFS) of 14 months and a median OS of 22 months (36).

Radioembolization has emerged as a valuable and increasingly utilized treatment option for patients with advanced GBC. Although it is typically offered as a palliative treatment for liver tumors, ongoing advances are broadening the range of treatment options for patients with advanced GBC and offer a promising avenue for future research and clinical applications.

Targeted therapy and immunotherapy

Advanced sequencing technologies, such as next-generation sequencing (NGS), whole exome sequencing, RNA sequencing and single-cell analysis, have enabled the identification of genetic and epigenetic features, as well as key molecules that could serve as potential therapeutic targets for GBC (3739). Kuipers et al (40) found that tumor protein p53 (TP53; ~57%), sonic hedgehog signaling molecule (~20%), E74-like ETS transcription factor 3 (~18.6%) and AT-rich interaction domain 1A (~14%) are among the most commonly mutated genes in GBC. Additionally, mothers against DPP homolog 4 (~13.1%), epidermal growth factor receptor (EGFR; also known as HER1/ERBB1; ~12%), erb-b2 receptor tyrosine kinase 2 (ERBB2; also known as HER2/NEU; ~10%), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α (~14.6%) and KRAS proto-oncogene GTPase (KRAS; ~10.3%) were also identified (4144). Ongoing clinical trials investigating targeted therapies and immunotherapy options for patients with GBC are summarized in Table II (4548).

Table II.

Main clinical trials of targeted therapy for gallbladder cancer.

Table II.

Main clinical trials of targeted therapy for gallbladder cancer.

NCT ID or first author, yearYearPhaseTargeted therapyTreatment regimensPrimary end pointStatus(Refs.)
NCT055123772022-IITP53Brigimadlin (BI 907828)NoneOngoing
Kim et al, 20202014-2017IIEGFR and VEGF pathwaysRegorafenibOS, 6.7 monthsComplete(45)
Lee et al, 20232020-2023IIERBB2 (human/neu)Trastuzumab + FOLFOX (95% CI, 16.7–46.3)ORR, 29.4%Complete(46)
Costello et al, 20142009-2012IPI3K/AKT/mTOR pathwayGemcitabine and cisplatinCR, 2 (7.4%); PR, 3 (11.1%)Complete(47)
Bekaii-Saab et al, 20232020-2022IIKRASG12CAdagrasibDOR, 5.3 months (95% CI, 2.8–7.3); PFS, 7.4 months (95% CI, 5.3–8.6)Complete(48)

[i] NCT ID, National Clinical Trial identification; OR, objective response; OS, overall survival; ORR, objective response rate; CR, complete response; PR, partial response; DOR, duration of response; PFS, progression-free survival; FOLFOX, 5-fluorouracil + leucovorin + oxaliplatin; TP53, tumor protein p53; EGFR, epidermal growth factor receptor; VEGF, vascular endothelial growth factor; ERBB2, erb-b2 receptor tyrosine kinase 2; KRAS, KRAS proto-oncogene GTPase.

TP53

Mutations in the TP53 tumor suppressor gene are present in ~50% of all human cancer cases. TP53 acts as a transcription factor that directly regulates the expression of ~500 genes and blocks their tumor suppressive functions by preventing them from binding to target DNA sequences. TP53 also upregulates genes involved in cellular processes, such as apoptosis, cell cycle arrest and senescence, which are key mechanisms that prevent tumorigenesis (4951). The frequency of TP53 mutations varies notably across different tumor types. TP53 mutations are present in >90% of ovarian cancer cases, whereas <15% of cases of acute myeloid leukemia exhibit TP53 mutations. This disparity suggests the possibility of tissue-specific factors influencing the loss of wild-type TP53 function or the gain of function in mutated p53 (52). The expression and accumulation levels of p53 increase from precancerous lesions (dysplasia) to aggressive GBC (5355). Detection of p53 protein upregulation can be a valuable diagnostic tool for accurate cancer identification. Stancu et al (56) found that p53 is upregulated in 30% of papillary regions, 30–50% of undifferentiated adenocarcinomas and 80–100% of highly differentiated adenocarcinomas, particularly in the context of chronic cholecystitis (56). In general, TP53 represents a promising therapeutic target, and with the advancement of p53-targeted therapies, personalized treatment plans based on specific p53 mutations are becoming increasingly feasible.

A phase IIa/IIb, open-label, single-arm, multicenter trial of brigimadlin (BI 907828) is currently ongoing in patients with locally advanced/metastatic, mouse double minute 2-amplified, TP53 wild-type biliary tract adenocarcinoma, pancreatic ductal adenocarcinoma and other selected solid tumors. The study aims to assess the efficacy of BI 907828 in treating cancers affecting the biliary tract, pancreas, lungs or bladder (57).

TP53 mutations have significant effects on several aspects of the immune system; they can promote immune evasion and cancer progression by regulating exosomes, growth factors, cytokines and chemokines (58). These mutations can alter the immune recognition of tumor cells and, through mechanisms such as reduced infiltration, decrease the cytotoxicity and activity of immune cells, thereby promoting the recruitment of immune-suppressive cells (59,60).

EGFR and vascular endothelial growth factor (VEGF) pathways

Both EGFR and VEGFR activate signaling pathways that regulate cell growth and proliferation (9,61). Additionally, the simultaneous inhibition of both EGFR and VEGFR may enhance antitumor effects (62). Studies on GBC have shown that overexpression of VEGF-A plays a crucial role in angiogenesis and serves as an independent prognostic factor associated with reduced survival (63). VEGF overexpression in GBC cells promotes angiogenesis, cell proliferation and metastasis (64,65). Up to 48% of patients with GBC exhibit elevated VEGF-A expression, potentially benefiting from anti-angiogenic drugs (63). Research has proved the efficacy of drugs targeting tumor angiogenesis via the VEGF/VEGFR pathway (66,67).

Bevacizumab is a humanized monoclonal antibody targeting VEGFA, while ramucirumab regulates VEGFR-2 specifically (68). Regorafenib, a novel oral multikinase inhibitor, blocks various vasogenic and stromal receptor tyrosine kinases (FGFR1, PDGFR-OMAN, TIE2, VEFR1, VEF2 and VVF3) and downstream cellular signaling pathways (RAS/LUR/MEK/ERK and PI3K/PTEN/AKT) (6971). In a multi-center, open-label, single-arm phase 2 trial evaluating regorafenib efficacy in refractory BTC, 33 patients treated for at least 3 weeks. The results showed a median PFS time of 3.9 months and an OS time of 6.7 months. The target response rate was 9.1%, with a disease control rate of 63.6% (45).

ERBB2

The receptor tyrosine protein kinase ERBB2, also known as HER2, is a member of the receptor tyrosine kinase EGFR family. Heterodimerization of this receptor with other members of the EGFR family leads to the self-phosphorylation of tyrosine residues in the heterodimeric cytoplasmic domain and initiates multiple signaling pathways, leading to cell proliferation and tumorigenesis (72). ERBB2 upregulation has been observed in several solid tumors, such as breast cancer, gastroesophageal junction cancer and BTC (7377). Nam et al (78) found that in patients with gallbladder cancer (N=21), the proportion of HER2-positive disease was 28.6%.

A phase 2 clinical trial, which was investigator-initiated, open-label, non-randomized and multi-institutional, evaluated the efficacy of treatment in patients aged ≥19 years with ERBB2-positive BTC (including intrahepatic cholangiocarcinoma, extrahepatic cholangiocarcinoma and GBC) that had progressed despite GEM and CDDP chemotherapy (79). The trial reported an overall response rate of 29.4% (95% CI, 16.7–46.3) and a disease control rate of 79.4% (95% CI, 62.9–89.9). The median PFS time was 5.1 months (95% CI, 3.6–6.7), while the median OS time was 10.7 months (95% CI, 7.9-not reached) (46).

PI3K/AKT/mTOR pathway

The PI3K-AKT-mTOR signaling pathway is a critical pathway involved in drug resistance and malignant processes in patients with solid cancer, with dysregulation potentially leading to various human cancers (7982). This pathway is increasingly being recognized as a promising target in cancer and related diseases, such as hepatocellular carcinoma and colon cancer (83). Abnormal activation of the PI3K-AKT -mTOR pathway has been observed in GBC and in other malignancies, such as prostate, breast and endometrial cancer (42). Activation of this pathway promotes increased cell proliferation and growth, and plays a key role in angiogenesis. The pathway is also involved in numerous cellular processes, including proliferation, invasion, migration, apoptosis and metabolism (8486). Targeting the PI3K/AKT/mTOR pathway in combination with chemotherapy or other targeted therapies can help inhibit tumor progression (87).

In a phase I trial, a chemotherapy regimen combining everolimus, an mTOR inhibitor, with GEM and CDDP resulted in stable disease in 6 out of 10 patients with BTC, suggesting a potential therapeutic benefit (47).

KRAS

KRAS, a member of the RAS oncogene family, was first identified in the 1980's by Barbacid (88) in a human bladder cancer cell line. This gene, also known as HRAS, is a homolog located on chromosome 11 (11p15.1–11p15.3) (88). KRAS is the best-known oncogene, the most common driver of numerous cancer types, with relatively high mutation rates in various cancers, such as non-small cell lung cancer, colorectal cancer and pancreatic ductal adenocarcinoma (89,90). Numerous studies have demonstrated the biochemical heterogeneity of KRAS mutations across various aspects, including intrinsic GTPase activity and affinities for effectors and binding sites. For instance, alterations in codons 12, 13 and 61 typically result in impaired intrinsic GTPase activity in KRAS, unlike the mutations KRAS G12D, KRAS G12C and KRAS G13D (91,92). The simultaneous activation of KRAS and the typical Wnt pathway induces BTC (93). Initial clinical reports for KRASG12C-mutated BTC showed a limited clinical response to sotorasib and GDC-6036 (48), and secondary mutations in the KRAS gene conferred acquired resistance to KRASG12C inhibitors. A phase II cohort of the KRYSTAL-1 study (ClinicalTrials.gov identifier: NCT03785249) was begun to evaluate adagrasib (600 mg orally twice daily) in patients with KRASG12C-mutated advanced solid tumors (excluding non-small cell lung cancer and colorectal cancer). As of October 1, 2022, 64 patients with KRASG12C-mutated solid tumors were enrolled and 63 patients treated (median follow-up, 16.8 months). Among 57 patients with measurable disease at baseline, the median duration of response was 5.3 months (95% CI, 2.8–7.3) and the median PFS time was 7.4 months (95% CI, 5.3–8.6) (48). KRAS mutant tumors exhibit significant heterogeneity, highlighting the urgent need for selective inhibitors that target specific KRAS mutations. Notable progress has been made in targeting KRAS, particularly KRASG12C. New approaches, including NMR-based fragment screening, drug teaming and computational drug design, have been employed to discover novel compounds that bind directly to KRAS (92). However, achieving effective KRAS targeting is still a considerable challenge. Such treatments are essential to effectively inhibit the function of various KRAS mutants, aligning with the goals of precision oncology (92).

Immune checkpoint inhibitors

Chemotherapy primarily delays tumor growth by inhibiting DNA replication, blocking the cell cycle and interfering with cellular metabolism (94). Additionally, some cytotoxic chemotherapy agents, such as anthracyclines and OXA, can induce immunogenic cell death and stimulate antitumor immune responses (95,96). Therefore, for chemotherapy-based immunotherapy with antibodies targeting programmed cell death protein 1/programmed death ligand 1 (anti-PD-1/PD-L1) may serve as a suitable partner to enhance the ability of chemotherapy to control tumor growth (97).

Currently, the common immune checkpoints of GBC include PD-1/PD-L1 and cytotoxic T-lymphocyte protein 4 (CTLA-4), and monoclonal antibodies targeting these targets have also been tested in clinical trials (98100). PD-1 serves as a well-known immunosuppressive checkpoint, whereas PD-L1 acts as its ligand (101). PD-L1 is a biomarker for patient prognosis and susceptibility to PD-1/PD-L1 inhibitors. PD-L1 is expressed mainly in tumor cells, tumor-infiltrating cells and antigen-presenting cells in a number of cancer types, such as breast and bone cancer (102). Numerous studies have affirmed the clinical importance of PD-1/PD-L1 antibodies in influencing the prognosis of human cancer (97,103105). Based on the immense success of clinical trials, ten anti-PD-1 antibodies (nivolumab, pembrolizumab, cemiplimab, sintilimab, camrelizumab, toripalimab, tislelizumab, zimberelimab, prolgolimab and dostarlimab) and three anti-PD-L1 antibodies (atezolizumab, durvalumab and avelumab) have been approved for the treatment of various types of cancer, including pancreatic and gastric cancer (106).

A multicenter phase II study involving 54 patients assessed the effectiveness and safety of nivolumab monotherapy in patients with advanced cholangiocarcinoma who had undergone first to third-line treatment. The study found that PD-L1 upregulation was significantly associated with worse OS times (hazard ratio, 1.58; 95% CI, 1.30–1.92; P<0.001) (107). However, serious challenges remain, such as limited beneficiaries, high drug resistance, lack of predictive and prognostic biomarkers, and treatment-related adverse reactions (108). Moreover, few predictive biomarkers can identify the types of patients who will benefit from treatment (109). Nevertheless, PD-1/PD-L1 immune checkpoint inhibitors have wide application prospects and clinical value for the treatment of cancer in humans.

CTLA-4, a receptor found on activated T cells, has been the focus of numerous clinical trials on combination therapies. The use of both CTLA-4 and PD-1 inhibitors has shown effectiveness in enhancing the response rates and survival of patients with various cancer types, including pancreatic and gastric cancer (110,111). However, combination therapy may also increase the risk of adverse events (112). A recent phase I clinical trial (NCT01938612) revealed that treatment with durvalumab and tremelimumab in patients with BTC led to a median OS time of 10.1 months (95% CI, 6.5–11.6) compared with 8.1 months (95% CI, 5.6–10.1) in patients with durvalumab monotherapy (113).

Adoptive immunotherapy

Immunotherapy has recently revolutionized cancer treatment and constitutes the fourth cornerstone of cancer therapy following surgery, RT and chemotherapy. To date, >700 clinical trials of CAR-T therapy have been registered at clinicaltrials.gov, with a number focusing on solid tumors. CAR-T cell immunotherapy has achieved marked success in the treatment of hematological malignancies. However, only six chimeric antigen receptor T-cell (CAR-T) cell products approved by the US Food and Drug Administration (FDA) for the treatment of R/R B cell malignancies, including tisagenlecleucel (Kymriah; Novartis), axicabtagene ciloleucel (Yescarta; Gilead), brexucabtagene autoleucel (Tecartus; Gilead), lisocabtagene maraleucel (Breyanzi; Bristol Myers Squibb), idecabtagene vicleucel (Abecma; Bristol Myers Squibb and Bluebird Bio), and ciltacabtagene autoleucel (Carvykti; Legend and Janssen) (114). There are currently six commercial CAR-T products that have been FDA approved for diseases such as B-acute lymphoblastic leukemia, large B-cell lymphoma, mantle cell lymphoma (MCL), follicular lymphoma, multiple myeloma and chronic lymphocytic leukemia/small lymphocytic lymphoma (115). In a single-arm, open-label, first-in-human phase I pilot study (NCT03159819), patients aged 18–70 years with confirmed CLDN18.2-positive advanced gastric or pancreatic cancer received one or more cycles of CAR-CLDN18.2 T cell infusion following lymphocyte pretreatment, until disease progression or the onset of intolerable toxicity. At a median follow-up of 14.3 months after infusion, the estimates for 6- and 12-month PFS rates were 69% (95% CI, 61–75) and 59% (95% CI, 51–66), respectively (116). Brexucabtagene autoleucel (brexu-cel) is an autologous CD19-directed CAR T-cell therapy approved for relapsed/refractory MCL. Patients who underwent leukapheresis between August 1, 2020, and December 31, 2021, at 16 US institutions, with an intent to manufacture commercial brexu-cel for relapsed/refractory MCL, were included. Of 189 patients who underwent leukapheresis, 168 (89%) received brexu-cel infusion. At a median follow-up of 14.3 months after infusion, the estimates for 6- and 12-month PFS rates were 69% (95% CI, 61–75) and 59% (95% CI, 51–66), respectively (117). However, due to the complexity of solid tumors and their unique anatomical locations, treating solid tumors with CAR-T cells presents numerous challenges, such as the lack of cancer-specific antigens, inefficient transport and migration of CAR-T cells to the tumor site. Consequently, the immunosuppressive tumor microenvironment (TME) and CAR-T therapy for solid tumors remain underdeveloped (114,118,119). As a result, CAR-T therapy for solid tumors is still in its developmental stages (120). However, clinical studies have demonstrated promising antitumor efficacy of CAR-T therapy, and the potential remains.

In recent years, various strategies have been developed to enhance the efficacy and safety of CAR-T cell therapies for solid tumors, primarily by addressing challenges posed by the unique characteristics of T cells and the TME. Companion diagnostics, such as immunohistochemistry and circulating tumor cell detection assays, can play a crucial role in improving the treatment of solid tumors with CAR-T cells (121).

Tumor vaccine

There are four types of cancer vaccines: Tumor- or immune cell-based vaccines, peptide-based vaccines, viral vector-based vaccines and nucleic acid-based vaccines (122). Vaccines based on nucleic acids (DNA or RNA) show great promise. mRNA cancer vaccines stand out due to their efficiency, safe management, rapid development potential and cost-effectiveness. Clinical trials have demonstrated positive results for over two dozen mRNA-based immunotherapies in treating solid tumors. Notably, a 3-year phase 1 follow-up from BioNTech's neoantigen individualized mRNA vaccine [autogene cevumeran (BNT122, RO7198457)] clinical study for patients after complete resection of pancreatic ductal adenocarcinoma has shown promising results (123).

Cancer vaccines are an effective treatment option as they stimulate a durable immune response that targets and eliminates tumor cells. However, the complexity of the immune system makes the development of effective cancer vaccines challenging. In particular, the heterogeneity of solid tumors may require different vaccinations. With the promising results from multiple clinical trials involving mRNA cancer vaccines against aggressive solid tumors, rapid advancements in mRNA vaccine technology can be anticipated for cancer immunotherapy in the near future (124).

Palliative treatment

The goal of palliative care for patients with GBC is to ensure unobstructed biliary drainage and provide nutritional support. Biliary drainage can be achieved using percutaneous catheterization or endoscopic biliary stenting to alleviate biliary obstruction, relieve liver cholestasis and enhance liver function. A multicenter retrospective analysis was performed of all cases of consecutive endoscopic ultraound-guided gallbladder drainage (EUS-GBD) with lumen-apposing metal stents (LAMSs) used as a rescue treatment for patients with distal malignant biliary obstruction (DMBO) in 14 Italian centers between June 2015 and June 2020. The study showed that EUS-GBD with LAMSs used as a rescue treatment for patients affected by DMBO represents a valuable option in terms of technical and clinical success rates, with an acceptable adverse event rate (5 patients; 10.4%) (125).

Tumors often compress the stomach or duodenum. In situations where the tumor compresses the stomach or duodenum, various interventions, such as nasoenteral nutrition tubes, gastrojejunal bypass therapy and peripheral nutrition support, can be employed to improve the patient's nutritional status (126). In addition to prolonging life, effective relief of symptoms is also of utmost importance for two reasons: i) Relief of physical suffering is desirable and can improve quality of life; and ii) effective symptom control translates into improved existential well-being (127). As advanced tumors often involve tumor spread or metastasis, a single treatment approach is often insufficient to achieve optimal treatment results. Therefore, palliative care also requires the collaboration of experts from multiple disciplinary fields to develop and implement individualized treatment programs aimed at improving patient outcomes and quality of life (128). At the same time, psychological intervention and symptomatic treatment should be carried out for patients. Psychosocial care for advanced cancer encompasses a wide range of interventions that help patients make life-changing decisions, relieve patient suffering, confront impending mortality and improve other patient outcomes (129). Cancer care guidelines also recommend distress screening for patients with cancer and the provision of psychological support when necessary, although a number of patients decline such support when offered.

Tondorf et al (130) conducted a prospective observational study to assess distress, the intention to utilize psycho-oncology support, and the acceptance of psycho-oncology services. Of the 333 patients assessed (mean age, 61 years; 55% male; 54% distress thermometer ≥5), 25% intended to use the psycho-oncology service (yes), 33% were ambivalent (maybe) and 42% reported no intention (no). Overall, 23% had attended the psycho-oncology service 4 months later.

Challenges and future prospects

The treatment of GBC, the most common malignancy of BTC, has progressed over the past 20 years. However, the treatment of advanced GBC remains challenging due to chemotherapy resistance and tumor heterogeneity due to mutations of genes such as KRAS and TP53. Given the improving understanding of the unique tumor biology of GBC, treatments for advanced GBC have changed markedly. The treatment options are no longer limited to single chemotherapy or local treatment, and the application of combination therapy can improve the prognosis of patients with advanced GBC. NGS of genes, related molecular genetic analysis and biomarker studies will aid the development of effective targets and drugs for GBC therapy, improve patient prognosis and improve therapeutic effectiveness.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Key Joint Project of Chongqing Health Commission and Science and Technology Bureau (grant no. 2024ZDXM007) and the Key Medical Research Project of Daping Hospital (grant no. ZXAIZD003).

Availability of data and materials

Not applicable.

Authors' contributions

GC conceived and designed the study, SRZ carried out the data collection and BZ wrote the manuscript. All authors contributed to the revision of the manuscript and have read and approved the final version. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Roa JC, García P, Kapoor VK, Maithel SK, Javle M and Koshiol J: Gallbladder cancer. Nat Rev Dis Primers. 8:692022. View Article : Google Scholar : PubMed/NCBI

2 

Song X, Hu Y, Li Y, Shao R, Liu F and Liu Y: Overview of current targeted therapy in gallbladder cancer. Signal Transduct Target Ther. 5:2302020. View Article : Google Scholar : PubMed/NCBI

3 

Zhao Y, Yang M, Feng J, Wang X and Liu Y: Advances in immunotherapy for biliary tract cancers. Chin Med J (Engl). 137:524–532. 2024. View Article : Google Scholar : PubMed/NCBI

4 

Schmidt MA, Marcano-Bonilla L and Roberts LR: Gallbladder cancer: Epidemiology and genetic risk associations. Chin Clin Oncol. 8(31)2019.PubMed/NCBI

5 

Lau CSM, Zywot A, Mahendraraj K and Chamberlain RS: Gallbladder carcinoma in the United States: A population based clinical outcomes study involving 22,343 patients from the surveillance, epidemiology, and end result database (1973–2013). HPB Surg. 2017:15328352017. View Article : Google Scholar : PubMed/NCBI

6 

Hickman L and Contreras C: Gallbladder cancer: Diagnosis, surgical management, and adjuvant therapies. Surg Clin North Am. 99:337–355. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Feo CF, Ginesu GC, Fancellu A, Perra T, Ninniri C, Deiana G, Scanu AM and Porcu A: Current management of incidental gallbladder cancer: A review. Int J Surg. 98:1062342022. View Article : Google Scholar : PubMed/NCBI

8 

Wang J, Ren M, Yu J, Hu M, Wang X, Ma W, Jiang X and Cui J: Single-cell RNA sequencing highlights the functional role of human endogenous retroviruses in gallbladder cancer. EBioMedicine. 85:1043192022. View Article : Google Scholar : PubMed/NCBI

9 

Shen H, He M, Lin R, Zhan M, Xu S, Huang X, Xu C, Chen W, Yao Y, Mohan M and Wang J: PLEK2 promotes gallbladder cancer invasion and metastasis through EGFR/CCL2 pathway. J Exp Clin Cancer Res. 38:2472019. View Article : Google Scholar : PubMed/NCBI

10 

Wacker F and Dewald C: Local and locoregional treatment of intrahepatic cholangiocarcinoma. Radiologe. 62:247–252. 2022.(In German). View Article : Google Scholar : PubMed/NCBI

11 

Scott A, Wong P and Melstrom LG: Surgery and hepatic artery infusion therapy for intrahepatic cholangiocarcinoma. Surgery. 174:113–115. 2023. View Article : Google Scholar : PubMed/NCBI

12 

Edeline J, Lamarca A, McNamara MG, Jacobs T, Hubner RA, Palmer D, Groot Koerkamp B, Johnson P, Guiu B and Valle JW: Locoregional therapies in patients with intrahepatic cholangiocarcinoma: A systematic review and pooled analysis. Cancer Treat Rev. 99:1022582021. View Article : Google Scholar : PubMed/NCBI

13 

Mondaca S, Yarmohammadi H and Kemeny NE: Regional Chemotherapy for Biliary Tract Tumors and Hepatocellular Carcinoma. Surg Oncol Clin N Am. 28:717–729. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Chang Y, Jeong SW, Young Jang J and Jae Kim Y: Recent updates of transarterial chemoembolilzation in hepatocellular carcinoma. Int J Mol Sci. 21:81652020. View Article : Google Scholar : PubMed/NCBI

15 

Ray CE Jr, Edwards A, Smith MT, Leong S, Kondo K, Gipson M, Rochon PJ, Gupta R, Messersmith W, Purcell T and Durham J: Metaanalysis of survival, complications, and imaging response following chemotherapy-based transarterial therapy inpatients with unresectable intrahepatic cholangiocarcinoma. J Vasc Interv Radiol. 24:1218–1226. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Larghi A, Rimbas M, Tringali A, Boškoski I, Rizzatti G and Costamagna G: Endoscopic radiofrequency biliary ablation treatment: A comprehensive review. Dig Endosc. 31:245–255. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Deufel CL, Furutani KM, Dahl RA, Grams MP, McLemore LB, Hallemeier CL, Neben-Wittich M, Martenson JA and Haddock MG: Technique for the administration of high-dose-rate brachytherapy to the bile duct using a nasobiliary catheter. Brachytherapy. 17:718–725. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Yin Z, Jin H, Ma T, Zhou Y, Yu M and Jian Z: A meta-analysis of long-term survival outcomes between Surgical resection and radiofrequency ablation in patients with a single hepatocellular carcinoma <2 cm (BCLC very early stage). Int J Surg. 56:61–67. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Gkika E, Hawkins MA, Grosu AL and Brunner TB: The evolving role of radiation therapy in the treatment of biliary tract cancer. Front Oncol. 10:6043872020. View Article : Google Scholar : PubMed/NCBI

20 

Franzese C, Bonu ML, Comito T, Clerici E, Loi M, Navarria P, Franceschini D, Pressiani T, Rimassa L and Scorsetti M: Stereotactic body radiotherapy in the management of oligometastatic and recurrent biliary tract cancer: Single-institution analysis of outcome and toxicity. J Cancer Res Clin Oncol. 146:2289–2297. 2020. View Article : Google Scholar : PubMed/NCBI

21 

Bisello S, Buwenge M, Palloni A, Autorino R, Cellini F, Macchia G, Deodato F, Cilla S, Brandi G, Tagliaferri L, et al: Radiotherapy or chemoradiation in unresectable biliary cancer: A retrospective study. Anticancer Res. 39:3095–3100. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Komiyama S, Takeda A, Tateishi Y, Tsurugai Y, Eriguchi T and Horita N: Comparison of stereotactic body radiotherapy and transcatheter arterial chemoembolization for hepatocellular carcinoma: Systematic review and meta-analysis. Radiother Oncol. 202:1106142025. View Article : Google Scholar : PubMed/NCBI

23 

Gkika E, Hallauer L, Kirste S, Adebahr S, Bartl N, Neeff HP, Fritsch R, Brass V, Nestle U, Grosu AL and Brunner TB: Stereotactic body radiotherapy (SBRT) for locally advanced intrahepatic and extrahepatic cholangiocarcinoma. BMC Cancer. 17:7812017. View Article : Google Scholar : PubMed/NCBI

24 

Salgado SM, Gaidhane M and Kahaleh M: Endoscopic palliation of malignant biliary strictures. World J Gastrointest Oncol. 8:240–247. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Sasaki T, Takeda T, Okamoto T, Ozaka M and Sasahira N: Chemotherapy for biliary tract cancer in 2021. J Clin Med. 10:3108–3122. 2021. View Article : Google Scholar : PubMed/NCBI

26 

Yang L, Wang H, Guo M, He M, Zhang W, Zhan M and Liu Y: ELF3 promotes gemcitabine resistance through PKMYT1/CDK1 signaling pathway in gallbladder cancer. Cell Oncol (Dordr). 46:1085–1095. 2023. View Article : Google Scholar : PubMed/NCBI

27 

Choi IS, Kim KH, Lee JH, Suh KJ and Kim JW, Park JH, Kim YJ, Kim JS, Kim JH and Kim JW: A randomised phase II study of oxaliplatin/5-FU (mFOLFOX) versus irinotecan/5-FU (mFOLFIRI) chemotherapy in locally advanced or metastatic biliary tract cancer refractory to first-line gemcitabine/cisplatin chemotherapy. Eur J Cancer. 154:288–295. 2021. View Article : Google Scholar : PubMed/NCBI

28 

Sharma A, Kalyan Mohanti B, Pal Chaudhary S, Sreenivas V, Kumar Sahoo R, Kumar Shukla N, Thulkar S, Pal S, Deo SV, Pathy S, et al: Modified gemcitabine and oxaliplatin or gemcitabine + cisplatin in unresectable gallbladder cancer: Results of a phase III randomised controlled trial. Eur J Cancer. 123:162–170. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Valle J, Wasan H, Palmer DH, Cunningham D, Anthoney A, Maraveyas A, Madhusudan S, Iveson T, Hughes S, Pereira SP, et al: Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. N Engl J Med. 362:1273–1281. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Morizane C, Okusaka T, Mizusawa J, Katayama H, Ueno M, Ikeda M, Ozaka M, Okano N, Sugimori K, Fukutomi A, et al: Combination gemcitabine plus S-1 versus gemcitabine plus cisplatin for advanced/recurrent biliary tract cancer: The FUGA-BT (JCOG1113) randomized phase III clinical trial. Ann Oncol. 30:1950–1958. 2019. View Article : Google Scholar : PubMed/NCBI

31 

Sharma A, Dwary AD, Mohanti BK, Deo SV, Pal S, Sreenivas V, Raina V, Shukla NK, Thulkar S, Garg P and Chaudhary SP: Best supportive care compared with chemotherapy for unresectable gallbladder cancer: A randomized controlled study. J Clin Oncol. 28:4581–4586. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Lai J, Yang S, Lin Z, Huang W, Li X, Li R, Tan J and Wang W: Update on chemoresistance mechanisms to first-line chemotherapy for gallbladder cancer and potential reversal strategies. Am J Clin Oncol. 46:131–141. 2023. View Article : Google Scholar : PubMed/NCBI

33 

Ben-Josef E, Guthrie KA, El-Khoueiry AB, Corless CL, Zalupski MM, Lowy AM, Thomas CR Jr, Alberts SR, Dawson LA, Micetich KC, et al: SWOG S0809: A phase II intergroup trial of adjuvant capecitabine and gemcitabine followed by radiotherapy and concurrent capecitabine in extrahepatic cholangiocarcinoma and gallbladder carcinoma. J Clin Oncol. 33:2617–2622. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Lamarca A, Palmer DH, Wasan HS, Ross PJ, Ma YT, Arora A, Falk S, Gillmore R, Wadsley J, Patel K, et al: Second-line FOLFOX chemotherapy versus active symptom control for advanced biliary tract cancer (ABC-06): A phase 3, open-label, randomized, controlled trial. Lancet Oncol. 22:690–701. 2021. View Article : Google Scholar : PubMed/NCBI

35 

Dodagoudar C, Doval DC, Mahanta A, Goel V, Upadhyay A, Goyal P, Talwar V, Singh S, John MC, Tiwari S and Patnaik N: FOLFOX-4 as second-line therapy after failure of gemcitabine and platinum combination in advanced gall bladder cancer patients. Jpn J Clin Oncol. 46:57–62. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Edeline J, Touchefeu Y, Guiu B, Farge O, Tougeron D, Baumgaertner I, Ayav A, Campillo-Gimenez B, Beuzit L, Pracht M, et al: Radioembolization plus chemotherapy for first-line treatment of locally advanced intrahepatic cholangiocarcinoma: A phase 2 clinical trial. JAMA Oncol. 6:51–59. 2020. View Article : Google Scholar : PubMed/NCBI

37 

Zhou Y, Yuan K, Yang Y, Ji Z, Zhou D, Ouyang J, Wang Z, Wang F, Liu C, Li Q, et al: Gallbladder cancer: Current and future treatment options. Front Pharmacol. 14:11836192023. View Article : Google Scholar : PubMed/NCBI

38 

Schmid S, Jochum W, Padberg B, Demmer I, Mertz KD, Joerger M, Britschgi C, Matter MS, Rothschild SI and Omlin A: How to read a next-generation sequencing report-what oncologists need to know. ESMO Open. 7:1005702022. View Article : Google Scholar : PubMed/NCBI

39 

McCombie WR, McPherson JD and Mardis ER: Next-generation sequencing technologies. Cold Spring Harb Perspect Med. 9:a0367982019. View Article : Google Scholar : PubMed/NCBI

40 

Kuipers H, de Bitter Tjjde Boer MT, van der Post RS, Nijkamp MW, de Reuver PR, Fehrmann RSN and Hoogwater FJH: Gallbladder cancer: Current insights in genetic alterations and their possible therapeutic implications. Cancers (Basel). 13:52572021. View Article : Google Scholar : PubMed/NCBI

41 

Javle M, Bekaii-Saab T, Jain A, Wang Y, Kelley RK, Wang K, Kang HC, Catenacci D, Ali S, Krishnan S, et al: Biliary cancer: Utility of next-generation sequencing for clinical management. Cancer. 122:3838–3847. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Roa I, Garcia H, Game A, de Toro G, de Aretxabala X and Javle M: Somatic mutations of PI3K in early and advanced gallbladder cancer: Additional options for an orphan cancer. J Mol Diagn. 18:388–394. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Dixit R, Pandey M, Tripathi SK, Dwivedi AN and Shukla VK: Comparative analysis of mutational profile of sonic hedgehog gene in gallbladder cancer. Dig Dis Sci. 62:708–714. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Yang P, Javle M, Pang F, Zhao W, Abdel-Wahab R, Chen X, Meric-Bernstam F, Chen H, Borad MJ, Liu Y, et al: Somatic genetic aberrations in gallbladder cancer: Comparison between Chinese and US patients. Hepatobiliary Surg Nutr. 8:604–614. 2019. View Article : Google Scholar : PubMed/NCBI

45 

Kim RD, Sanoff HK, Poklepovic AS, Soares H, Kim J, Lyu J, Liu Y, Nixon AB and Kim DW: A multi-institutional phase 2 trial of regorafenib in refractory advanced biliary tract cancer. Cancer. 126:3464–3470. 2020. View Article : Google Scholar : PubMed/NCBI

46 

Lee CK, Chon HJ, Cheon J, Lee MA, Im HS, Jang JS, Kim MH, Park S, Kang B, Hong M, et al: Trastuzumab plus FOLFOX for HER2-positive biliary tract cancer refractory to gemcitabine and cisplatin: a multi-institutional phase 2 trial of the Korean Cancer Study Group (KCSG-HB19-14). Lancet Gastroenterol Hepatol. 8:56–65. 2023. View Article : Google Scholar : PubMed/NCBI

47 

Costello BA, Borad MJ, Qi Y, Kim GP, Northfelt DW, Erlichman C and Alberts SR: Phase I trial of everolimus, gemcitabine and cisplatin in patients with solid tumors. Investig. New Drugs. 32:710–716. 2014. View Article : Google Scholar

48 

Bekaii-Saab TS, Yaeger R, Spira AI, Pelster MS, Sabari JK, Hafez N, Barve M, Velastegui K, Yan X, Shetty A, et al: Adagrasib in advanced solid tumors harboring a KRASG12C mutation. J Clin Oncol. 41:4097–4106. 2023. View Article : Google Scholar : PubMed/NCBI

49 

Kastenhuber ER and Lowe SW: Putting p53 in Context. Cell. 170:1062–1078. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Vousden KH and Lane DP: p53 in health and disease. Nat Rev Mol Cell Biol. 8:275–283. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Levine AJ: p53: 800 million years of evolution and 40 years of discovery. Nat Rev Cancer. 20:471–480. 2020. View Article : Google Scholar : PubMed/NCBI

52 

Kennedy MC and Lowe SW: Mutant p53: It's not all one and the same. Cell Death Differ. 29:983–987. 2022. View Article : Google Scholar : PubMed/NCBI

53 

Ajiki T, Onoyama H, Yamamoto M, Asaka K, Fujimori T, Maeda S and Saitoh Y: p53 protein expression and prognosis in gallbladder carcinoma and premalignant lesions. Hepatogastroenterology. 43:521–526. 1996.PubMed/NCBI

54 

Zhao C, Yang ZY, Zhang J, Li O, Liu SL, Cai C, Shu YJ, Pan LJ, Gong W and Dong P: Inhibition of XPO1 with KPT-330 induces autophagy-dependent apoptosis in gallbladder cancer by activating the p53/mTOR pathway. J Transl Med. 20:4342022. View Article : Google Scholar : PubMed/NCBI

55 

Nepal C, Zhu B, O'Rourke CJ, Bhatt DK, Lee D, Song L, Wang D, Van Dyke AL, Choo-Wosoba H, Liu Z, et al: Integrative molecular characterisation of gallbladder cancer reveals micro-environment-associated subtypes. J Hepatol. 74:1132–1144. 2021. View Article : Google Scholar : PubMed/NCBI

56 

Stancu M, Caruntu ID, Sajin M, Giuşca S, Badescu A and Dobrescu G: Immunohistochemical markers in the study of gallbladder premalignant lesions and cancer. Rev Med Chir Soc Med Nat Iasi. 111:734–743. 2007.PubMed/NCBI

57 

National Library of Medicine, . Brightline-2: A Study to Test Whether Brigimadlin (BI 907828) Helps People With Cancer in the Biliary Tract, Pancreas, Lung or Bladder. https://ClinicalTrials.gov/show/NCT05512377

58 

Pilley S, Rodriguez TA and Vousden KH: Mutant p53 in cell-cell interactions. Genes Dev. 35:433–448. 2021. View Article : Google Scholar : PubMed/NCBI

59 

Agupitan AD, Neeson P, Williams S, Howitt J, Haupt S and Haupt Y: p53: A guardian of immunity becomes its saboteur through mutation. Int J Mol Sci. 21:34522020. View Article : Google Scholar : PubMed/NCBI

60 

Efe G, Rustgi AK and Prives C: p53 at the crossroads of tumor immunity. Nat Cancer. 5:983–995. 2024. View Article : Google Scholar : PubMed/NCBI

61 

Guo XF, Zhang YY, Kang J, Dou QH and Zhu XF: A bispecific decoy receptor VEGFR-EGFR/Fc binding EGF-like ligands and VEGF shows potent antitumor efficacy. J Drug Target. 30:302–312. 2022. View Article : Google Scholar : PubMed/NCBI

62 

Hung MS, Chen IC, Lin PY, Lung JH, Li YC, Lin YC, Yang CT and Tsai YH: Epidermal growth factor receptor mutation enhances expression of vascular endothelial growth factor in lung cancer. Oncol Lett. 12:4598–4604. 2016. View Article : Google Scholar : PubMed/NCBI

63 

Singh P, Jain SL, Sakhuja P and Agarwal A: Expression of VEGF-A, HER2/neu, and KRAS in gall bladder carcinoma and their correlation with clinico-pathological parameters. Indian J Pathol Microbiol. 64:687–692. 2021. View Article : Google Scholar : PubMed/NCBI

64 

Xu D, Li J, Jiang F, Cai K and Ren G: The effect and mechanism of vascular endothelial growth factor (VEGF) on tumor angiogenesis in gallbladder carcinoma. Iran J Public Health. 48:713–721. 2019.PubMed/NCBI

65 

De Lorenzo S, Garajova I, Stefanini B and Tovoli F: Targeted therapies for gallbladder cancer: An overview of agents in preclinical and clinical development. Expert Opin Investig Drugs. 30:759–772. 2021. View Article : Google Scholar : PubMed/NCBI

66 

Mabeta P and Steenkamp V: The VEGF/VEGFR axis revisited: Implications for cancer therapy. Int J Mol Sci. 23:155852022. View Article : Google Scholar : PubMed/NCBI

67 

Patel SA, Nilsson MB, Le X, Cascone T, Jain RK and Heymach JV: Molecular mechanisms and future implications of VEGF/VEGFR in cancer therapy. Clin Cancer Res. 29:30–39. 2023. View Article : Google Scholar : PubMed/NCBI

68 

Ahamed A, Samanta A, Alam SSM, Mir SA, Jamil Z, Ali S and Hoque M: Nonsynonymous mutations in VEGF receptor binding domain alter the efficacy of bevacizumab treatment. J Cell Biochem. 125:e305152024. View Article : Google Scholar : PubMed/NCBI

69 

Papadimitriou M and Papadimitriou CA: Antiangiogenic tyrosine kinase inhibitors in metastatic colorectal cancer: Focusing on regorafenib. Anticancer Res. 41:567–582. 2021. View Article : Google Scholar : PubMed/NCBI

70 

Yan H, Wu W, Hu Y, Li J, Xu J, Chen X, Xu Z, Yang X, Yang B, He Q and Luo P: Regorafenib inhibits EphA2 phosphorylation and leads to liver damage via the ERK/MDM2/p53 axis. Nat Commun. 14:27562023. View Article : Google Scholar : PubMed/NCBI

71 

Sahin IH, Tan E and Kim R: Regorafenib, an investigational agent for the treatment of cholangiocarcinoma. Expert Opin Investig Drugs. 30:333–341. 2021. View Article : Google Scholar : PubMed/NCBI

72 

Yarden Y and Sliwkowski MX: Untangling the ErbB signaling network. Nat Rev Mol Cell Biol. 2:127–137. 2001. View Article : Google Scholar : PubMed/NCBI

73 

Li M, Liu F, Zhang F, Zhou W, Jiang X, Yang Y, Qu K, Wang Y, Ma Q, Wang T, et al: Genomic ERBB2/ERBB3 mutations promote PD-L1-mediated immune escape in gallbladder cancer: A whole-exome sequencing analysis. Gut. 68:1024–1033. 2019. View Article : Google Scholar : PubMed/NCBI

74 

Augustin JE, Soussan P and Bass AJ: Targeting the complexity of ERBB2 biology in gastroesophageal carcinoma. Ann Oncol. 33:1134–1148. 2022. View Article : Google Scholar : PubMed/NCBI

75 

Fernandez AI, Liu M, Bellizzi A, Brock J, Fadare O, Hanley K, Harigopal M, Jorns JM, Kuba MG, Ly A, et al: Examination of Low ERBB2 Protein Expression in Breast Cancer Tissue. JAMA Oncol. 8:1–4. 2022. View Article : Google Scholar : PubMed/NCBI

76 

Kam AE, Masood A and Shroff RT: Current and emerging therapies for advanced biliary tract cancers. Lancet Gastroenterol Hepatol. 6:956–969. 2021. View Article : Google Scholar : PubMed/NCBI

77 

Merters J and Lamarca A: Integrating cytotoxic, targeted and immune therapies for cholangiocarcinoma. J Hepatol. 78:652–657. 2023. View Article : Google Scholar : PubMed/NCBI

78 

Nam AR, Kim JW, Cha Y, Ha H, Park JE, Bang JH, Jin MH, Lee KH, Kim TY, Han SW, et al: Therapeutic implication of HER2 in advanced biliary tract cancer. Oncotarget. 7:58007–58021. 2016. View Article : Google Scholar : PubMed/NCBI

79 

National Library of Medicine, . Trastuzumab-pkrb Combined With Modified FOLFOX-6 in Biliary Tract Cancer Patients Progressed on First Line Therapy. https://ClinicalTrials.gov/show/NCT04722133

80 

Peng Y, Wang Y, Zhou C, Mei W and Zeng C: PI3K/Akt/mTOR pathway and its role in cancer therapeutics: Are we making headway? Front Oncol. 12:8191282022. View Article : Google Scholar : PubMed/NCBI

81 

Lavacchi D, Caliman E, Rossi G, Buttitta E, Botteri C, Fancelli S, Pellegrini E, Roviello G, Pillozzi S and Antonuzzo L: Ivosidenib in IDH1-mutated cholangiocarcinoma: Clinical evaluation and future directions. Pharmacol Ther. 237:1081702022. View Article : Google Scholar : PubMed/NCBI

82 

Wang J, Jiang W, Liu S, Shi K, Zhang Y, Chen Y, Shan J, Wang Y, Xu X, Li C and Li X: Exosome-derived miR-182-5p promoted cholangiocarcinoma progression and vasculogenesis by regulating ADK/SEMA5a/PI3K pathway. Liver Int. 44:370–388. 2024. View Article : Google Scholar : PubMed/NCBI

83 

Narayanankutty A: PI3K/akt/mTOR pathway as a therapeutic target for colorectal cancer: A review of preclinical and clinical evidence Curr. Drug Targets. 20:1217–1226. 2019. View Article : Google Scholar : PubMed/NCBI

84 

Rosenberg L, Yoon CH, Sharma G, Bertagnolli MM and Cho NL: Sorafenib inhibits proliferation and invasion in desmoid-derived cells by targeting Ras/MEK/ERK and PI3K/Akt/mTOR pathways. Carcinogenesis. 39:681–688. 2018. View Article : Google Scholar : PubMed/NCBI

85 

Cai C, Dang W, Liu S, Huang L, Li Y, Li G, Yan S, Jiang C, Song X, Hu Y and Gu J: Anthrax toxin receptor 1/tumor endothelial marker 8 promotes gastric cancer progression through activation of the PI3K/AKT/mTOR signaling pathway. Cancer Sci. 111:1132–1145. 2020. View Article : Google Scholar : PubMed/NCBI

86 

Liu H, Liu C, Wang M, Sun D, Zhu P, Zhang P, Tan X and Shi G: Tanshinone IIA affects the malignant growth of Cholangiocarcinoma cells by inhibiting the PI3K-Akt-mTOR pathway. Sci Rep. 11:192682021. View Article : Google Scholar : PubMed/NCBI

87 

Zhong L, Li Y, Xiong L, Wang W, Wu M, Yuan T, Yang W, Tian C, Miao Z, Wang T and Yang S: Small molecules in targeted cancer therapy: Advances, challenges, and future perspectives. Signal Transduct Target Ther. 6:2012021. View Article : Google Scholar : PubMed/NCBI

88 

Barbacid M: ras genes. Annu Rev Biochem. 56:779–827. 1987. View Article : Google Scholar : PubMed/NCBI

89 

Hallin J, Bowcut V, Calinisan A, Briere DM, Hargis L, Engstrom LD, Laguer J, Medwid J, Vanderpool D, Lifset E, et al: Anti-tumor efficacy of a potent and selective non-covalent KRASG12D inhibitor. Nat Med. 8:2171–2182. 2022. View Article : Google Scholar : PubMed/NCBI

90 

Zhu C, Guan X, Zhang X, Luan X, Song Z, Cheng X, Zhang W and Qin JJ: Targeting KRAS mutant cancers: From druggable therapy to drug resistance. Mol Cancer. 21:1592022. View Article : Google Scholar : PubMed/NCBI

91 

Hunter JC, Manandhar A, Carrasco MA, Gurbani D, Gondi S and Westover KD: Biochemical and structural analysis of common cancer-associated KRAS mutations. Mol Cancer Res. 13:1325–1335. 2015. View Article : Google Scholar : PubMed/NCBI

92 

Huang L, Guo Z, Wang F and Fu L: KRAS mutation: From undruggable to druggable in cancer. Signal Transduct Target Ther. 6:3862021. View Article : Google Scholar : PubMed/NCBI

93 

Nagao M, Fukuda A, Omatsu M, Namikawa M, Sono M, Fukunaga Y, Masuda T, Araki O, Yoshikawa T, Ogawa S, et al: Concurrent activation of Kras and canonical Wnt signaling induces premalignant lesions that progress to extrahepatic biliary cancer in mice. Cancer Res. 82:1803–1817. 2022. View Article : Google Scholar : PubMed/NCBI

94 

Gotwals P, Cameron S, Cipolletta D, Cremasco V, Crystal A, Hewes B, Mueller B, Quaratino S, Sabatos-Peyton C, Petruzzelli L, et al: Prospects for combining targeted and conventional cancer therapy with immunotherapy. Nat Rev Cancer. 17:286–301. 2017. View Article : Google Scholar : PubMed/NCBI

95 

Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini JL, Castedo M, Mignot G, Panaretakis T, Casares N, et al: Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med. 13:54–61. 2007. View Article : Google Scholar : PubMed/NCBI

96 

Zhu H, Shan Y, Ge K, Lu J, Kong W and Jia C: Oxaliplatin induces immunogenic cell death in hepatocellular carcinoma cells and synergizes with immune checkpoint blockade therapy. Cell Oncol (Dordr). 43:1203–1214. 2020. View Article : Google Scholar : PubMed/NCBI

97 

Yi M, Zheng X, Niu M, Zhu S, Ge H and Wu K: Combination strategies with PD-1/PD-L1 blockade: current advances and future directions. Mol Cancer. 21:282022. View Article : Google Scholar : PubMed/NCBI

98 

Wang L, Tang K, Li X and Lu W: Anti-PD-1-based immunotherapy plus lenvatinib to treat advanced gallbladder cancer in the elderly: A case series and review of current literature. J Cancer Res Clin Oncol. 149:941–950. 2023. View Article : Google Scholar : PubMed/NCBI

99 

Patel SP, Guadarrama E, Chae YK, Dennis MJ, Powers BC, Liao CY, Ferri WA Jr, George TJ, Sharon E, Ryan CW, et al: SWOG 1609 cohort 48: Anti-CTLA-4 and anti-PD-1 for advanced gallbladder cancer. Cancer. 130:2918–2927. 2024. View Article : Google Scholar : PubMed/NCBI

100 

Zheng Q, Wu C, Ye H, Xu Z, Ji Y, Rao J, Lu L, Zhu Y and Cheng F: Analysis of the efficacy and prognostic factors of PD-1 inhibitors in advanced gallbladder cancer. Ann Transl Med. 9:15682021. View Article : Google Scholar : PubMed/NCBI

101 

Jalili-Nik M, Soltani A, Mashkani B, Rafatpanah H and Hashemy SI: PD-1 and PD-L1 inhibitors foster the progression of adult T-cell Leukemia/Lymphoma. Int Immunopharmacol. 98:1078702021. View Article : Google Scholar : PubMed/NCBI

102 

Zuo H and Wan Y: Inhibition of myeloid PD-L1 suppresses osteoclastogenesis and cancer bone metastasis. Cancer Gene Ther. 29:1342–1354. 2022. View Article : Google Scholar : PubMed/NCBI

103 

Luke JJ, Rutkowski P, Queirolo P, Del Vecchio M, Mackiewicz J, Chiarion-Sileni V, de la Cruz Merino L, Khattak MA, Schadendorf D, Long GV, et al: Pembrolizumab versus placebo as adjuvant therapy in completely resected stage IIB or IIC melanoma (KEYNOTE-716): A randomized, double-blind, phase 3 trial. Lancet. 399:1718–1729. 2022. View Article : Google Scholar : PubMed/NCBI

104 

Wu M, Huang Q, Xie Y, Wu X, Ma H, Zhang Y and Xia Y: Improvement of the anticancer efficacy of PD-1/PD-L1 blockade via combination therapy and PD-L1 regulation. J Hematol Oncol. 15:242022. View Article : Google Scholar : PubMed/NCBI

105 

Chu X, Tian W, Wang Z, Zhang J and Zhou R: Co-inhibition of TIGIT and PD-1/PD-L1 in cancer immunotherapy: Mechanisms and clinical trials. Mol Cancer. 22:932023. View Article : Google Scholar : PubMed/NCBI

106 

Tang Q, Chen Y, Li X, Long S, Shi Y, Yu Y, Wu W, Han L and Wang S: The role of PD-1/PD-L1 and application of immune-checkpoint inhibitors in human cancers. Front Immunol. 13:9644422022. View Article : Google Scholar : PubMed/NCBI

107 

Xie Q, Wang L and Zheng S: Prognostic and clinicopathological significance of PD-L1 in patients with cholangiocarcinoma: A meta-analysis. Dis Markers. 2020:18179312020. View Article : Google Scholar : PubMed/NCBI

108 

Xia L, Liu Y and Wang Y: PD-1/PD-L1 blockade therapy in advanced non-small cell lung cancer: Current status and future directions. Oncologist. 24 (Suppl 1):S31–S41. 2019. View Article : Google Scholar : PubMed/NCBI

109 

Aguiar PN Jr, De Mello RA, Hall P, Tadokoro H and Lima Lopes G: PD-L1 expression as a predictive biomarker in advanced non-small cell lung cancer: Updated survival data. Immunotherapy. 9:499–506. 2017. View Article : Google Scholar : PubMed/NCBI

110 

Zhang H, Dai Z, Wu W, Wang Z, Zhang N, Zhang L, Zeng WJ, Liu Z and Cheng Q: Regulatory mechanisms of immune checkpoints PD-L1 and CTLA-4 in cancer. J Exp Clin Cancer Res. 40:1842021. View Article : Google Scholar : PubMed/NCBI

111 

Sznol M and Melero I: Revisiting anti-CTLA-4 antibodies in combination with PD-1 blockade for cancer immunotherapy. Ann Oncol. 32:295–297. 2021. View Article : Google Scholar : PubMed/NCBI

112 

Rotte A: Combination of CTLA-4 and PD-1 blockers for treatment of cancer. J Exp Clin Cancer Res. 38:2552019. View Article : Google Scholar : PubMed/NCBI

113 

Doki Y, Ueno M, Hsu CH, Oh DY, Park K, Yamamoto N, Ioka T, Hara H, Hayama M, Nii M, et al: Tolerability and efficacy of durvalumab, either as monotherapy or in combination with tremelimumab, in patients from Asia with advanced biliary tract, esophageal, or head-and-neck cancer. Cancer Med. 11:2550–2560. 2022. View Article : Google Scholar : PubMed/NCBI

114 

Zhang X, Zhu L, Zhang H, Chen S and Xiao Y: CAR-T cell therapy in hematological malignancies: Current opportunities and challenges. Front Immunol. 13:9271532022. View Article : Google Scholar : PubMed/NCBI

115 

Goyco Vera D, Waghela H, Nuh M, Pan J and Lulla P: Approved CAR-T therapies have reproducible efficacy and safety in clinical practice. Hum Vaccin Immunother. 20:23785432024. View Article : Google Scholar : PubMed/NCBI

116 

National Library of Medicine, . Clinical Study of CAR-CLD18 T Cells in Patients With Advanced Gastric Adenocarcinoma and Pancreatic Adenocarcinoma. https://ClinicalTrials.gov/show/NCT03159819

117 

Wang Y, Jain P, Locke FL, Maurer MJ, Frank MJ, Munoz JL, Dahiya S, Beitinjaneh AM, Jacobs MT, Mcguirk JP, et al: Brexucabtagene autoleucel for relapsed or refractory mantle cell lymphoma in standard-of-care practice: Results from the US lymphoma CAR T consortium. J Clin Oncol. 41:2594–2606. 2023. View Article : Google Scholar : PubMed/NCBI

118 

Depil S, Duchateau P, Grupp SA, Mufti G and Poirot L: ‘Off-the-shelf’ allogeneic CAR T cells: Development and challenges. Nat Rev Drug Discov. 19:185–199. 2020. View Article : Google Scholar : PubMed/NCBI

119 

Sterner RC and Sterner RM: CAR-T cell therapy: Current limitations and potential strategies. Blood Cancer J. 11:692021. View Article : Google Scholar : PubMed/NCBI

120 

Pan K, Farrukh H, Chittepu VCSR, Xu H, Pan CX and Zhu Z: CAR race to cancer immunotherapy: from CAR T, CAR NK to CAR macrophage therapy. J Exp Clin Cancer Res. 41:1192022. View Article : Google Scholar : PubMed/NCBI

121 

Ma S, Li X, Wang X, Cheng L, Li Z, Zhang C, Ye Z and Qian Q: Current progress in CAR-T cell therapy for solid tumors. Int J Biol Sci. 15:2548–2560. 2019. View Article : Google Scholar : PubMed/NCBI

122 

Faghfuri E, Pourfarzi F, Faghfouri AH, Abdoli Shadbad M, Hajiasgharzadeh K and Baradaran B: Recent developments of RNA-based vaccines in cancer immunotherapy. Expert Opin Biol Ther. 21:201–218. 2020. View Article : Google Scholar : PubMed/NCBI

123 

Malacopol AT and Holst PJ: Cancer vaccines: Recent insights and future directions. Int J Mol Sci. 25:112562024. View Article : Google Scholar : PubMed/NCBI

124 

Miao L, Zhang Y and Huang L: mRNA vaccine for cancer immunotherapy. Mol Cancer. 20:412021. View Article : Google Scholar : PubMed/NCBI

125 

Binda C, Anderloni A, Fugazza A, Amato A, de Nucci G, Redaelli A, Di Mitri R, Cugia L, Pollino V, Macchiarelli R, et al: EUS-guided gallbladder drainage using a lumen-apposing metal stent as rescue treatment for malignant distal biliary obstruction: A large multicenter experience. Gastrointest Endosc. 98:765–773. 2023. View Article : Google Scholar : PubMed/NCBI

126 

Benson AB, D'Angelica MI, Abbott DE, Anaya DA, Anders R, Are C, Bachini M, Borad M, Brown D, Burgoyne A, et al: Hepatobiliary cancers, version 2.2021, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 19:541–565. 2021. View Article : Google Scholar : PubMed/NCBI

127 

Strang P: Palliative oncology and palliative care. Mol Oncol. 16:3399–3409. 2022. View Article : Google Scholar : PubMed/NCBI

128 

Beasley AM, Bakitas MA, Ivankova N and Shirey MR: Evolution and conceptual foundations of nonhospice palliative care. West J Nurs Res. 41:1347–1369. 2019. View Article : Google Scholar : PubMed/NCBI

129 

Kozlov E, Niknejad B and Reid MC: Palliative care gaps in providing psychological treatment: A review of the current state of research in multidisciplinary palliative care. Am J Hosp Palliat Care. 35:505–510. 2018. View Article : Google Scholar : PubMed/NCBI

130 

Tondorf T, Grossert A, Rothschild SI, Koller MT, Rochlitz C, Kiss A, Schaefert R, Meinlschmidt G, Hunziker S and Zwahlen D: Focusing on cancer patients' intentions to use psychooncological support: A longitudinal, mixed-methods study. Psychooncology. 27:1656–1663. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2025
Volume 30 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhou B, Zhang S and Chen G: Development and challenges in the treatment of advanced gallbladder cancer (Review). Oncol Lett 30: 382, 2025.
APA
Zhou, B., Zhang, S., & Chen, G. (2025). Development and challenges in the treatment of advanced gallbladder cancer (Review). Oncology Letters, 30, 382. https://doi.org/10.3892/ol.2025.15128
MLA
Zhou, B., Zhang, S., Chen, G."Development and challenges in the treatment of advanced gallbladder cancer (Review)". Oncology Letters 30.2 (2025): 382.
Chicago
Zhou, B., Zhang, S., Chen, G."Development and challenges in the treatment of advanced gallbladder cancer (Review)". Oncology Letters 30, no. 2 (2025): 382. https://doi.org/10.3892/ol.2025.15128