1,043
Views
0
CrossRef citations to date
0
Altmetric
Review

Treatment resistance in pancreatic and biliary tract cancer: molecular and clinical pharmacology perspectives

, , , , , & show all
Pages 323-347 | Received 20 Nov 2023, Accepted 12 Feb 2024, Published online: 15 Mar 2024

ABSTRACT

Introduction

Treatment resistance poses a significant obstacle in oncology, especially in biliary tract cancer (BTC) and pancreatic cancer (PC). Current therapeutic options include chemotherapy, targeted therapy, and immunotherapy. Resistance to these treatments may arise due to diverse molecular mechanisms, such as genetic and epigenetic modifications, altered drug metabolism and efflux, and changes in the tumor microenvironment. Identifying and overcoming these mechanisms is a major focus of research: strategies being explored include combination therapies, modulation of the tumor microenvironment, and personalized approaches.

Areas covered

We provide a current overview and discussion of the most relevant mechanisms of resistance to chemotherapy, target therapy, and immunotherapy in both BTC and PC. Furthermore, we compare the different strategies that are being implemented to overcome these obstacles.

Expert opinion

So far there is no unified theory on drug resistance and progress is limited. To overcome this issue, individualized patient approaches, possibly through liquid biopsies or single-cell transcriptome studies, are suggested, along with the potential use of artificial intelligence, to guide effective treatment strategies. Furthermore, we provide insights into what we consider the most promising areas of research, and we speculate on the future of managing treatment resistance to improve patient outcomes.

1. Introduction and current therapeutic options

The term, biliary tract cancers (BTCs), refers to malignancies that arise in the bile ducts, including intrahepatic bile ducts, extrahepatic bile ducts, and the gallbladder. Overall, BTCs are relatively rare compared to other cancers, accounting for approximately 3% of all gastrointestinal malignancies, but with various incidence rates around the world [Citation1]. In the United States, the estimated annual incidence of BTC is around 2,500 to 3,000 cases [Citation2]. The risk factors for BTC include chronic inflammation of the bile ducts (such as in primary sclerosing cholangitis), gallstones, liver fluke infections, and genetic alterations [Citation1].

Pancreatic cancer (PC) is a highly aggressive malignancy that arises in the tissues of the pancreas. It is more common than BTC, being the 12th most common cancer worldwide and accounting for approximately 2% of all new cancer cases [Citation3]. In the United States, PC is the 10th most common cancer, and it is the third leading cause of cancer-related deaths, having an estimated annual incidence of around 60,000 cases. The risk factors for PC include smoking, obesity, chronic pancreatitis, diabetes, family history of PC, as well as some genetic alterations [Citation4].

1.1. Treatment algorithm in biliary tract cancer (BTC)

Despite the rarity of this disease, recent years have witnessed a substantial shift in the treatment paradigm of advanced BTC, with the introduction of both immunotherapy and target therapies (). Following the results of the phase-3 TOPAZ-1 trial, standard first-line treatment has become the combination of immune-chemotherapy with anti-programmed death-ligand 1 (PD-L1) durvalumab plus cisplatin – gemcitabine (GEM) [Citation5]. While the overall survival (OS) did not exhibit a substantial numerical increase (12.9 vs 11.3 months at the latest interim analysis), around 26% of patients in the immune-chemotherapy arm had a continued response after 1 year vs only 15% in the chemotherapy arm, showing how a long-lasting response can be obtained more frequently with immuno-chemotherapy [Citation6]. Similar OS results have been observed with the combination of anti PD-1 pembrolizumab plus cisplatin-GEM, with an estimated ongoing response rate at 24 months of 18% [Citation7].

Table 1. Treatment regimens in BTC.

Second-line treatment is now divided according to the presence of targetable mutations. Fibroblast growth factor receptor 2 (FGFR2) rearrangements have three Food and Drug Administration (FDA)-approved target drugs, pemigatinib, futibatinib, and infigratinib [Citation8–11]. Although efficacy varies with each drug, OS is overall significantly longer (up to 21.7 months with futibatinib), and duration of response can range from 5 months with infigratinib, up to 9.1 and 9.7 months with pemigatinib and futibatinib, respectively. IDH1 mutations can be targeted using FDA-approved ivosidenib, based on a small progression-free survival (PFS) gain against placebo (2.7 vs 1.4 months) and PFS rates at 12 months of 22% vs 0%, although overall response rate (ORR) was only 2% [Citation12]. Anti-Her2 therapy with zanidatamab was given FDA Breakthrough Approval in 2020 and the recent results of the HERIZON-BTC-01 trial are encouraging, with an ORR of 41% and PFS of 5.5 months [Citation13]. Notwithstanding the agnostic therapies available for alteration such as neurotrophic tyrosine receptor kinase (NTRK), BRAF, RET, and high microsatellite instability (MSI-H), many other drugs are in the early stages of testing in BTC. This suggests the potential for an expansion in approved targeted treatments for BTC in the near future [Citation14,Citation15] ().

Second-line standard of care treatment for patients with no targetable mutations is still chemotherapy, with no clear preferred treatments. One of the most common regimens, oxaliplatin–5-fluorouracil is associated with a small but statistically significant OS gain when compared to active symptom control (6.2 vs 5.3 months), but with poor ORR (5%) and disease control rate (33%), indicating high levels of primary resistance [Citation16]. A novel option is the use of liposomal-irinotecan +5-fluorouracil, associated with PFS ranging from 2.7 to 7.1 months, and ORR of around 15% [Citation17,Citation18]. Several chemotherapy drugs have been experimented within second or subsequent lines of therapy, yet none has demonstrated a clear advantage over the others [Citation19] ().

1.2. Treatment algorithm in pancreatic cancer (PC)

Chemotherapy with conventional anticancer agents is still the first-line treatment standard in metastatic PC (). The most commonly used regimens are FOLFIRINOX (5-fluorouracil + irinotecan + oxaliplatin) or GEM + nab-paclitaxel, both with an OS of less than 1 year (11 and 8.5 months respectively), ORR between 31.6% and 23%, and PFS rates at 12 months of 12% and 16% [Citation20,Citation21]. A similar 11.1 months OS result has been observed with the NALIRIFOX regimen (5-fluorouracil, liposomal irinotecan, oxaliplatin), although with a better PFS rate at 12 months of 27% vs 14% of the GEM + nab-paclitaxel combination [Citation22].

Table 2. Treatment regimens in PC.

When it comes to second or subsequent lines of therapy, the foundation remains chemotherapy-based, with many options available and no clear advantage of one over the others. Thus, treatment is usually personalized according to patients’ performance status, expected toxicities, and previous treatments. OS is usually between 3 and 9 months, and ORR varies between 10% and 20% for combination treatments to less than 10 for the monotherapies arm [Citation23–26] ().

Regarding novel approaches, target therapies have not yielded significant success in the context of PC. There is, however, an exception for maintenance therapy with poly(ADP-ribose) polymerase (PARP) inhibitor, olaparib, in the setting of BRCA1–2 germ-line mutated patients who have not progressed during first-line platinum-based chemotherapy: this therapy was associated with longer PFS (7.4 vs 3.8 months in the placebo arm) and a doubled PFS rate at 12 months (33.7 vs 14.5%), however, this advantage did not translate into an OS gain [Citation27]. Epidermal growth factor receptor (EGFR) inhibitor erlotinib has been tested alongside GEM vs GEM monotherapy, obtaining a 3 weeks gain in OS and similar ORR, thus this combo did not see much application in clinical practice [Citation28]. Although the rate of PCs with genomic alterations susceptible to agnostic therapies is very low, Tyrosine Kinase Inhibitor (TKI) treatment for NTRK fusion-positive cases and immunotherapy for cases with MSI-H can sometimes be applied [Citation29–31]. Many other targets are or have been tested, but none of them are being used outside of clinical trials [Citation32] ().

2. Molecular mechanisms of chemoresistance

In the field of medical research, gaining a deep understanding of the intricate molecular mechanisms underlying chemoresistance is of the utmost importance. Within the intricate web of cellular processes, various molecular players collaborate to orchestrate resistance, diminishing the efficacy of conventional treatments [Citation33]. Understanding these mechanisms holds great significance in the development of precision therapies that can overcome chemoresistance.

2.1. Chemoresistance in BTC

Despite advances in diagnostic techniques and therapeutic modalities, the prognosis for patients with BTC remains bleak, primarily due to the high incidence of chemoresistance. Here, we review the current knowledge regarding the molecular mechanisms contributing to chemoresistance in BTC ().

Figure 1. Molecular mechanisms of chemoresistance in BTC. chemoresistance in biliary tract cancer is mainly caused by epigenetic modifications, genetic mutations, hypoxic tumor microenvironment, great metastatic capacity of tumor cells and deregulation of metabolic pathways induced by chemotherapeutic agents, including changes in drug entry and exit transporters and changes in enzymes involved in drug effects. This is supported by the effective reduction of current cytotoxic drugs. Created with Biorender.com.

Figure 1. Molecular mechanisms of chemoresistance in BTC. chemoresistance in biliary tract cancer is mainly caused by epigenetic modifications, genetic mutations, hypoxic tumor microenvironment, great metastatic capacity of tumor cells and deregulation of metabolic pathways induced by chemotherapeutic agents, including changes in drug entry and exit transporters and changes in enzymes involved in drug effects. This is supported by the effective reduction of current cytotoxic drugs. Created with Biorender.com.

2.1.1. Genetic alterations

Genetic alterations play a pivotal role in the development of chemoresistance to various chemotherapeutic agents. However, preclinical studies regarding the role of different genetic aberrations in chemoresistance and their ‘targeting’ in BTC models are limited [Citation34] ().

A study in both intra- and extra-hepatic cholangiocarcinoma (ICC and ECC) cells demonstrated that enhancement of expression of the tumor suppressor gene TP53, resulted in the up-regulation of p21 and a reduction in cell proliferation [Citation35]. Furthermore, these models showed up-regulation of pro-tumoral proteins FAS, BAX, TYMP, and CES2, coupled with down-regulation of DHFR, RRM1, and BIRC5. These modifications were accompanied by increased sensitivity to antitumor drugs, particularly platinated drugs. Similar results were observed in ICC cells KKU-100 and KKU-M214, tested for their sensitivity to 5-fluorouracil, doxorubicin, and GEM [Citation36]. Enhanced chemosensitivity to all these anticancer drugs was attributed to the activation of p53-mediated cell death. Indeed, enhanced susceptibility to chemotherapeutic agents by the antioxidant/detoxifying enzyme NAD(P)H-quinone oxidoreductase-1 was abolished by knockdown of TP53, hinting to an important role of p53 and its alterations in the development of chemoresistance.

Dysregulation of oncogenes, such as the KRAS, might also contribute to chemoresistance by promoting cell survival and inhibiting apoptosis (). Gain-of-function mutations in KRAS are present in approximately 45–55% of ICC and 10–15% of ECC. Furthermore, one study showed that BRAF, an important downstream effector of KRAS, was mutated in up to 22% of ICC cases [Citation37].

Of note, Peng et al. (2023) investigated the correlation between TP53 and KRAS in cholangiocarcinoma (CCA) patients, showing that these alterations were significantly associated with shorter PFS [Citation38]. Thus, aberrations of TP53 and KRAS could serve as predictive indicators of chemoresistance and of an unfavorable prognosis in CCA patients ().

Interestingly, a recent study showed that the tyrosine phosphatase SHP2 directly governs the activity of the transcription regulator protein YAP by dephosphorylating it and establishing an association between reduced SHP2 phosphatase activity and chemoresistance in CCA cells, even in the context of RAS/RAF mutations [Citation39]. Cell lines characterized by low SHP2 expression and elevated phosphorylated form of YAP were resistant to GEM and cisplatin. Moreover, the marked chemoresistance of xenografts with genetically deleted SHP2 was overcome by using an inhibitor directed against YAP target genes, such as the antiapoptotic regulator MCL1.

Regarding relatively common mutations such as FGFR alterations, although data suggest a positive prognostic role, no correlation with response to first-line therapy has been found [Citation40,Citation41]. Conversely, alterations in DNA repair genes, such as BRCA1 and BRCA2, have been linked to higher sensitivity to platinum-based chemotherapy (). Kim et al. investigated the correlation between DNA repair gene mutations and the clinical response to platinum-based chemotherapy in patients with BTC and found that BRCA mutations were significantly associated with PFS at the multivariate analysis (HR 0.150, 95% CI: 0.034–0.655, p = 0.012) [Citation42]. This study demonstrated that BRCA mutations might have a role as predictive biomarkers for first-line platinum-based chemotherapy in patients with advanced BTC. Of note, a study in three CCA cell lines (QBC939, HuH28 and TFK-1), showed that the radiosensitivity of CCA cells was enhanced by PARP inhibitor olaparib, inducing DNA lesions and apoptosis [Citation43]. These findings hint to a strong relationship between gene alteration and response to therapy and spark the debate on overcoming resistance to traditional anticancer drugs by using synergistic combinations with novel targeted drugs ().

2.1.2. Epigenetic modifications

Epigenetic modifications, including DNA methylation, histone modifications, and non-coding RNA expression, have emerged as critical regulators of BTC pathogenesis ().

CCA tissue exhibits a significant decrease in DNA hydroxymethylation compared to non-tumor tissue [Citation44]. Additionally, hyper-methylation is observed in several gene promoters related to Wnt signaling, including the P16INK4a gene, found in up to 83% of CCA cases. P16INK4a inhibits the interaction between cyclin D1 and cyclin-dependent kinase 4 (CDK4). Methylation of the promoter halts P16INK4a activity, allowing CDK4 to bind to cyclin D1, initiating the cell’s entry into the S phase. The methylation of the CpG island is the primary factor leading to P16INK4a inactivation [Citation45]. Treatment with CDK4/6 inhibitors in 15 CCA cell lines resulted in decreased number of cells in S-phase and induced senescence. The efficacy of this treatment was further confirmed in spheroids and patient-derived xenografts [Citation46].

Furthermore, previous studies have highlighted the involvement of miRNAs in chemoresistance [Citation47] (). For example, Meng et al. demonstrated that miR-200b and miR-21 play roles in sensitivity to GEM, with their inhibition significantly increasing GEM cytotoxicity and apoptotic effects in CCA cell lines [Citation48]. Inhibition of miR-200b correlated with increased expression of protein phosphatase non-receptor type 12, impacting cell proliferation and differentiation, while inhibition of miR-21 led to decreased expression of its direct target, PTEN, influencing the PI-3 kinase pathway and promoting cell survival.

A study on the expression of 2555 miRNAs in CCA cells observed deregulation of 137 and 14 miRNAs in GEM-treated HuCCT-1 and TKKK cell lines, respectively, in comparison to their untreated controls [Citation49]. Specifically, miR-664b-3p, miR-3651, and miR-6087 exhibited increased expression in HuCCT-1 cell lines but were downregulated in TKKK cell lines, suggesting a potential role for these miRNAs in influencing the sensitivity of CCA cells to GEM [Citation49].

More recently, a high-throughput screening of 997 locked nucleic acid miRNA inhibitors in 6 CCA cell lines treated with cisplatin and GEM revealed that miR-1249 inhibition enhanced chemotherapy sensitivity across all tested cells [Citation50]. miR-1249 was found to be upregulated in CD133+ cells from human BTC stem cell niches and in chemo-resistant CCLP cells. Knockout of miR-1249 resulted in impaired expansion and enrichment of CD133+ subclones, reduced expression of cancer stem cell markers, and increased chemosensitivity. In xenograft models, miR-1249 knockout led to tumor shrinkage after exposure to weekly cisplatin and GEM, while wild-type models exhibited stable disease over treatment. Moreover, overexpression of miR-1249 was present in 41% of human BTCs cases, suggesting its potential both as biomarker of chemoresistance and as a potential target.

2.1.3. Tumor microenvironment (TME)

TME plays a crucial role in chemoresistance by providing a supportive niche for cancer cells (). Stromal cells, including cancer-associated fibroblasts (CAFs) and immune cells, secrete various factors, such as Transforming Growth Factor beta (TGF-β) and interleukins, that promote tumor growth and survival [Citation51] (). These factors can activate signaling pathways, such as PI3K/AKT and NF-κB, which contribute to chemoresistance [Citation52]. In particular, a recent study by Obata et al. (2023) demonstrated that this activation contributes to GEM resistance in biliary tract cancer cells, promoting cell survival and suppressing apoptosis [Citation53]. The study proposed miR-451a as a potential therapeutic target in gallbladder cancer (GBC), because it significantly impeded cell proliferation, induced apoptosis, and mitigated chemoresistant features, including epithelial–mesenchymal transition (EMT), in both GBC and GEM-resistant GBC. This effect was likely mediated through the negative regulation of the phosphatidylinositol 3-kinase/AKT pathway, achieved in part by directly downregulating macrophage migration inhibitory factor [Citation53].

In a separate study conducted by Yang et al., it was demonstrated that the NF-κB pathway plays a crucial role in the emergence of chemoresistance in biliary tract cancer (BTC) cells. GEM-resistant gallbladder cancer (GBC) cells exhibited low expression of KRAS and inactivation of AKT/ERK signaling. Conversely, in the same resistant cells the p70S6K, p38MAPK, and NF-κB signaling pathways were activated [Citation54].

Moreover, the hypoxic and nutrient-deprived conditions within TME can induce a quiescent state in cancer cells, rendering them less susceptible to chemotherapy [Citation55] (). Interestingly, the hypoxia-induced gene 2-oxoglutarate 5-dioxygenase 2 (PLOD2), was upregulated in BTC cells resistant to GEM, which were also characterized by low expression of epithelial markers and high expression of mesenchymal markers. The use of siRNA to downregulate PLOD2 led to reduction of chemoresistance, restoration of epithelial markers, and decrease of mesenchymal markers. In resected BTC samples, PLOD2 expression showed a significant correlation with lymph node metastasis and stage of the disease [Citation55]. Moreover, patients with high PLOD2 expression exhibited significantly lower recurrence-free survival and OS rates. Considering these findings, PLOD2 could be explored as a potential prognostic biomarker and therapeutic target for overcoming chemoresistance.

Collectively, we highlight the well-studied multifaceted nature of chemoresistance in BTC, urging further exploration of targeted therapies and combination strategies to enhance treatment efficacy and improve patient outcomes in this challenging disease.

2.2. Chemoresistance in PC

PC stands as one of the most formidable challenges in modern oncology, characterized by its aggressive nature and limited treatment success [Citation56,Citation57]. While significant strides have been made in understanding the molecular underpinnings of this malignancy, therapeutic progress has been impeded by the common inherent and acquired chemoresistance [Citation58]. This review seeks to provide an overview of the existing knowledge surrounding the molecular mechanisms implicated in chemoresistance in PC ().

Figure 2. Molecular mechanisms of chemoresistance in PC. chemoresistance in pancreatic cancer is mainly contributed by the epithelial–mesenchymal transition phenotype, genetic modifications, the hypoxic tumor microenvironment, the great metastatic capacity of tumor cells and the deregulation of metabolic pathways induced by chemotherapeutic agents, including changes in drug entry and exit transporters and changes in enzymes involved in drug effects. This is supported by the factors of an absence of symptoms that allow early detection and diagnosis, as well as the effective reduction of current cytotoxic drugs. Created with Biorender.com.

Figure 2. Molecular mechanisms of chemoresistance in PC. chemoresistance in pancreatic cancer is mainly contributed by the epithelial–mesenchymal transition phenotype, genetic modifications, the hypoxic tumor microenvironment, the great metastatic capacity of tumor cells and the deregulation of metabolic pathways induced by chemotherapeutic agents, including changes in drug entry and exit transporters and changes in enzymes involved in drug effects. This is supported by the factors of an absence of symptoms that allow early detection and diagnosis, as well as the effective reduction of current cytotoxic drugs. Created with Biorender.com.

2.2.1. Genetic alterations

Genetic alterations play a crucial role in the development of chemoresistance in PC. For instance, mutations in KRAS, which are detected in more than 90% of PC patients, have been associated with resistance to multiple chemotherapeutic agents (). Certain studies have also suggested that targeting KRAS may lead to the attenuation of chemoresistance and improve the therapeutic response in PC patients. Recent preclinical studies have shown that the use of SHP2 inhibitors (SHP099), MEK inhibitors (cobimetinib), and KRAS-G12C inhibitors (sotorasib), can enhance the efficacy of chemotherapeutic agents in PC cells carrying KRAS mutations, as well as an acceptable safety profile in pretreated patients with KRAS-mutated advanced PC [Citation59–61].

Alterations in TP53, have also been identified as key factors contributing to this resistance (). Pan and colleagues demonstrated that the loss of p53 function has been associated with aggressive tumor phenotypes, chemoresistance, and poor prognosis in PC patients, involving several mechanisms, such as dysregulation of cell cycle arrest, DNA damage repair, apoptosis, and autophagy [Citation62].

Mutations that impact genes responsible for repairing DNA damage, such as BRCA1/2, ATM, and PALB2, have been extensively studied for their role in response to chemotherapy [Citation63,Citation64] (). These genes encode important proteins involved in homology‐directed repair (HDR), which is a system for accurately fixing DNA double-strand breaks (DSB) [Citation65]. When there are mutations that disrupt the function of these genes, HDR is impaired, and cells rely more on error-prone repair mechanisms like non-homologous end joining and other DSB repair systems [Citation66]. In 2018, Blair et al. found that a group of PC patients with BRCA1/2 mutations had better outcomes when treated with platinum-based adjuvant chemotherapy compared to non-platinum-based adjuvant chemotherapy or nothing [Citation67]. Additionally, a retrospective study on 262 patients, of which 50 had HDR mutations, showed that HDR-mutated patients had better PFS when treated with first-line platinum therapy [Citation68]. Furthermore, a recent phase II study demonstrated that GEM + cisplatin treatment is effective against PCs with both BRCA1/2 and/or PALB2 mutations [Citation69].

2.2.2. Epigenetic modifications

Several epigenetic modifications, such as DNA methylation, histone acetylation and deacetylation, as well as histone methylation have been implicated in chemoresistance in PC. Most of these studies have demonstrated that disrupting DNA methylation homeostasis is a key factor in the development of human cancer. Furthermore, these studies have contributed to the realization that alterations in methylation patterns play a crucial role in distinguishing tumor cells and rendering them resistant to chemotherapy [Citation70]. The abnormal methylation patterns in PC have been extensively described [Citation71] ().

Aberrant expression of noncoding RNAs, such as microRNAs and long noncoding RNAs, can also impact the response to chemotherapy as they can act as oncogenes or tumor suppressor genes [Citation72–74] (). Hong and colleagues found that miR-21, miR-27a, miRNA-146a, miRNA-196a, and miRNA200a were the most upregulated miRNAs, while miR-20a, miR-96, and miR-217 were significantly downregulated in PC tissues [Citation75,Citation76]. This suggests that some miRNAs could promote oncogenic processes while others have tumor-suppressive effects. Several studies showed the role of miR-21 in chemoresistance and poor prognosis [Citation77–79], and the involvement of other miRNAs has been recently reviewed by Vahabi and collaborators [Citation80].

2.2.3. Epithelial–mesenchymal transition (EMT)

EMT is a critical process involved in the aggressive nature of cancers, leading to invasion, metastasis, and drug resistance [Citation81] (). El Amrani et al. conducted a study revealing that GEM induces EMT-like changes in PC cells [Citation82]. These changes are mediated by the extracellular regulated kinase (ERK)-zinc finger E-box binding homeobox 1 (ZEB1) pathway. Inhibition of ERK1/2 phosphorylation or ZEB1 expression leads to a reduction in chemoresistance and invasion of GEM-resistant (GR) PANC-1 and MIAPaCa-2 cells. Additionally, GEM induced overexpression of CD44, CD24, and CD326 in GR cells compared to sensitive cells. Further studies have identified that AMPK-related kinase 5 and upregulation of glycolysis contribute to GEM resistance through EMT mechanisms.

Kuwada et al. demonstrated that the induction of EMT in PANC-1 and BxPC-3 cells by tumor associated macrophages (TAMs) render them more migratory and thereby promote growth and chemoresistance [Citation83].

Lastly, EMT is closely linked to the modulation of the TGF-β pathway which is a key regulator of autophagy and apoptosis [Citation84]. The TGFβ-miR200-MIG6 pathway orchestrates a kinase switch linked to epithelial–mesenchymal transition (EMT), resulting in resistance to EGFR inhibitors [Citation85]. Noteworthy the antipsychotic drug brexpiprazole is able to overcome osimertinib resistance in both lung cancer and PC by suppressing survivin protein. This action subsequently inhibits TGF-β/SMAD signaling, preventing EMT [Citation86,Citation87]. The regulation of hMENA isoforms by TGF-β1 is pivotal in TGF-β1-induced EMT, presenting potential targets for the development of innovative prognostic and therapeutic strategies in PC [Citation88].

2.2.4. Altered drug influx and efflux

Human equilibrative nucleoside transporter-1 (hENT-1 or SLC29A1) has been shown to play a crucial role in the intracellular accumulation of nucleoside-based chemotherapeutic agents in PC cells, especially GEM [Citation89,Citation90] (). Previous studies reported that the downregulation/inhibition of hENT-1 resulted in cell chemoresistance to GEM and a retrospective analysis of the phase-3 clinical trials RTOG-9704 and ESPAC-1/3 supported the role of hENT1 as a predictive biomarker of GEM efficacy, with patients with high hENT-1 expression having a significantly longer OS [Citation91–93]. Of note, this association was missing in patients treated with 5-FU, suggesting a predictive and not prognostic role for hENT-1.

Similarly, another transport called dENT-2 (SLC29A2) has been identified in various PC cell lines, although with decreased expression compared to hENT-1 [Citation94]. It has been shown that h-ENT2 expression and its mRNA levels decrease after treatment with GEM in resistant cell lines [Citation95]. Expanding on these findings, deactivation of both hENT-2 and hENT-1 results in diminished GEM uptake and sensitivity.

Efflux is the active process through which cells pump drugs out of their cytoplasm to avoid drug buildup. Thus, the expression of efflux pumps like ATP-binding cassette (ABC) multi-drug transporters like ABCB1 have been found to play a role in drug resistance, drug distribution, and toxicity [Citation96] (). For instance, in the context of irinotecan, ABCB1 plays a role in the cellular uptake of both the prodrug and its active metabolite SN-38. However, the ABCB1 1236C >T variant markedly reduces irinotecan clearance, the ABCC2 3972T >C variant is linked to toxicity, and cells over-expressing ABCG2 are recognized as resistant to both irinotecan and SN-38 [Citation97].

Chen and collaborators reported a significantly lower promoter methylation level of the AB transporter family (ABCB1, ABCC, and ABCG2) in a GEM-resistant cell line (SW1990/GZ) compared to primary cells [Citation98]. Another study indicated a gradual decrease in the promoter methylation level of ABCB1, ABCC, and ABCG2 during the establishment of GEM resistance in PANC-1 and BxPC-3 cells [Citation99]. This provides evidence of the theoretical feasibility of predicting multi-drug resistance (MDR) of PC through independent indicators such as the promoter methylation level of ABCB1, ABCC and ABCG2. However, clinical trials evaluating the predictive role of these transporters did not translate into successful clinical application.

Other interesting studies evaluated the expression and activity of drug-metabolizing enzymes. Recently, a study conducted by Yada et al. investigated the role of cytochrome P450 enzymes in chemoresistance of PC, showing that mRNA expression of cytochrome-P450 was upregulated in a concentration-dependent manner following GEM treatment. Moreover, the sensitivity to GEM increased with the use of a cytochrome-P450 inhibitor, indicating that this enzyme may be related to GEM resistance in PC [Citation100].

The combination of nab-paclitaxel and GEM is another primary treatment option for patients diagnosed with advanced or metastatic PC [Citation21]. Notably, the efflux of paclitaxel is facilitated by phosphorylated glycoprotein [Citation101] and a recent study showed its key role in the resistance of SUIT02, PANC-1, and PaTu-T cells [Citation102]. The glycoprotein known as secreted protein acidic and rich in cysteine (SPARC) has been extensively studied as a potential biomarker for assessing the activity of nab-paclitaxel, correlating it with PC cell proliferation and metastasis [Citation103]. An initial immunohistochemical study conducted on 36 patients revealed a dose-dependent correlation between higher levels of SPARC expression and improved OS, however this was not conformed in following studies [Citation104]. Furthermore, nab-paclitaxel has exhibited the ability to inactivate cytidine deaminase, the enzyme which mediates the conversion of GEM into its inactive form, thus resulting in increased levels of GEM and a higher response rate in KPC models [Citation105].

2.2.5. Tumor microenvironment

Approximately 80% of PC tumor mass is constituted by stroma comprising cellular components (stellate cells, fibroblasts, endothelial and immune cells) and an extracellular matrix (ECM). Tumor stroma contributes to tumorigenesis by promoting invasion and enhancing tumoral angiogenesis, however the new vessels are often defective, explaining the hypoxic environment often observed in TME [Citation106]. In addition, the stroma creates physical barriers that limit drug penetration and promotes drug resistance, by activating signaling pathways that are involved in cell proliferation [Citation107,Citation108] (). Various growth factors (such as TGF-β, VEGF, CTGF, HGF, FGF), matricellular proteins, metalloproteinases, tissue inhibitors of metalloproteinases, and cytokines have been documented to impact drug effectiveness.

Sonic hedgehog (Shh), a soluble ligand, over-expressed by neoplastic cells in PC, drives formation of a fibroblast-rich desmoplastic stroma. Surprisingly, Shh-deficient tumors are more aggressive and exhibit undifferentiated histology, increased vascularity, and heightened proliferation features [Citation109]. Together, these data demonstrate that some components of the tumor stroma can also act to restrain tumor growth and should not be inhibited.

Some stromal cells, including CAFs and immune cells, can play an active role in promoting chemoresistance. They provide cancer cells with survival signals, activate signaling pathways that facilitate drug efflux, and secrete cytokines that hinder apoptosis [Citation110,Citation111]. Lastly, the ECM, composed of proteoglycans, hyaluronic acid (HA), and collagen, plays a crucial role in influencing interstitial fluid pressure and blood vessel distribution, contributing to hypoperfusion, hypoxia, and altered cancer cell metabolism. This, in turn, reduces drug delivery and activity [Citation112,Citation113] ().

In summary, PC presents one of the most formidable challenges in oncology, marked by its aggressive nature and limited treatment success, largely attributed to the pervasive issue of chemoresistance. This comprehensive review delves into the intricate molecular mechanisms underlying chemoresistance in PC, highlighting genetic alterations, epigenetic modifications, EMT, altered drug influx and efflux, and the TME as key contributors.

2.3. Overcoming chemoresistance

As seen before, the mechanisms underlying chemoresistance are complex, numerous, and often intertwined. Despite these difficulties, several attempts have been made to try to overcome chemoresistance in both BTC and PC ().

Table 3. Overcoming chemoresistance in BTC and PC.

Given the genetic alterations associated with chemoresistance, one approach is to combine standard chemotherapy with drugs directed against genes involved in chemoresistance, and this subject is explored in the dedicated chapter on target therapy ().

Figure 3. Mechanisms of resistance to target therapy and strategies to overcome them. there are three main mechanisms underlying resistance to targeted therapies: phenotypic transformations, on-target alterations and off-target alterations. Currently, attempts are being made to overcome this barrier by developing new drugs against common resistance mutations, combination of different treatments and personalized treatments based on liquid or tissue biopsy. Created with Biorender.com.

Figure 3. Mechanisms of resistance to target therapy and strategies to overcome them. there are three main mechanisms underlying resistance to targeted therapies: phenotypic transformations, on-target alterations and off-target alterations. Currently, attempts are being made to overcome this barrier by developing new drugs against common resistance mutations, combination of different treatments and personalized treatments based on liquid or tissue biopsy. Created with Biorender.com.

Regarding epigenetic changes that can lead to chemoresistance, Kurdistani et al. analyzed the role of histone modifications in GEM resistant PC [Citation72]. The researchers evaluated the effects of histone deacetylase inhibitors (HDACi) on PC cell lines and patient-derived xenograft models. They found that treatment with the HDACi vorinostat sensitized cells to GEM, resulting in decreased cell viability and increased apoptosis. Analysis of histone modifications revealed that treatment with vorinostat led to increased acetylation of histones H3 and H4, as well as decreased methylation of histone H3 lysine 27, which is associated with gene silencing. Furthermore, combination treatment with vorinostat and GEM resulted in improved survival in PC xenografts compared to either treatment alone. A neoadjuvant phase 1 trial with the combination of vorinostat plus multi TKI sorafenib and chemoradiotherapy demonstrated good tolerability and encouraging antitumoral activity [Citation114]. This suggests that modulation of histone modifications through HDACi holds potential for overcoming GEM-resistance in PC. Furthermore, DNA methyltransferase inhibitors have also being explored as epigenetic therapy: a phase 1 trial including both PC and BTC of guadecitabine and durvalumab was associated with a good tolerability profile and although the efficacy was limited, a group of patients had prolonged benefit, indicating the need for better biomarkers of response for this class of drugs [Citation115].

Epigenetic changes can also take the form of alternation in miRNA, thus several studies have explored the idea of miRNA silencing or the use of miRNA mimics [Citation116]. A new drug targeting the chemokine receptor CXCR4 along with miR-210 in CCA cell lines showed that this drug reversed the hypoxia-induced drug resistance and increased sensitivity to treatment with GEM plus cisplatin [Citation117]. Overall treatment with miRNA has been widely explored in the preclinical setting, however a phase-1 trial on a drug targeting miR-34a in solid tumors including PC and CCA [Citation118], showed that the treatment was associated with high infusion related adverse events.

Efforts to address phenotypic changes associated with chemoresistance due to EMT have been undertaken by targeting crucial pathways, albeit with limited success. For instance, drugs aimed at Notch, a pathway implicated in the emergence of CD44-positive PC stem cells and EMT in preclinical models, yielded unsatisfactory outcomes [Citation119]. In a phase-1/2 clinical trial, the monoclonal antibody against Notch2/Notch3 (tarextumab) failed to enhance overall survival in combination with GEM and nab-paclitaxel. Moreover, the gamma-secretase inhibitor MK0752, when combined with GEM, resulted in severe adverse effects, causing a high rate of patient withdrawal [Citation120,Citation121] ().

Recent studies have indicated that metformin, a medication used for diabetes, enhances the antiproliferative effects of PC cells by inhibiting EMT. This effect might potentially improve the overall prognosis of PC patients. In fact, therapeutic exposure to metformin was associated with reduced morbidity and mortality rates, but modestly increased short-term survival rates without affecting long-term survival rates in PC patients [Citation122].

Another important aspect of both PC and BTC is the TME and its role in favoring chemoresistance (). A therapeutic option may involve reversing the protumorigenic signals of CAFs or transforming them into a non-CAFs phenotype ‘re-educating’ CAFs to adopt a non-tumor-associated state [Citation123,Citation124]. One such approach involves the use of epigenetic modulating agents like the demethylating agent 5-Azacytidine (5-AZA). The administration of 5-AZA can transform an aggressive PC model into a less aggressive and more drug-sensitive phenotype. This transformation has been observed in vivo, with reduced tumor growth following engraftment of treated transformed cells. Additionally, when combined with GEM, 5-AZA exhibits a noticeable inhibitory effect on the growth of GEM-resistant PC cells [Citation125]. A phase 2 trial with azacitidine and anti PD-1 pembrolizumab as second-line treatment in PC did not show improved PFS, however one patient had a notably long survival and maintained response, indicating the need for predictive biomarkers [Citation126].

The stromal components in PC have been targeted using Hedgehog pathway inhibitors, such as IPI-926 or recombinant pegylated HA enzyme PEGPH20, however with unsatisfactory clinical outcomes [Citation127,Citation128]. A randomized phase-2 trial combining PEGPH20 with GEM and nab-paclitaxel in patients with high levels of HA only met the secondary endpoint of PFS and had to be temporarily halted due to increased thrombosis [Citation128]. Similarly, a randomized phase II trial investigating frontline PEGPH20 combined with FOLFIRINOX in a non-biomarker-selected population was terminated early due to lack of effectiveness in the interim analysis [Citation129]. Disappointing results have also been observed with inhibitors targeting the JAK2/STAT3 pathway, such as ruxolitinib, which did not show promising results in a phase-3 study when combined with capecitabine [Citation130,Citation131] ().

The first-line standard of care for more than 10 years before immunotherapy was included in the management of BTC was GEM with cisplatin, which was based on the groundbreaking phase III ABC-02 study that was published in 2010 [Citation132]. The doublet regimen achieved a median progression-free survival (mPFS) of 8.0 months and median overall survival (mOS) of 11.7 months, a statistically significant improvement compared to 5.0 months and 8.1 months for GEM alone. Patients were randomized to receive either GEM/cisplatin or GEM monotherapy. The objective response rate (ORR) was 26.1% and the disease control rate (DCR) was 81.4% for the doublet arm.

Treatment alternatives for those without modifiable changes include 5-FU/LV with oxaliplatin (5-FU/LV plus FOLFOX) and 5-FU/LV plus nanoliposomal (nal-)irinotecan. FOLFOX produced an ORR of 4.9% in the phase III ABC-06 study, resulting in a substantial but clinically negligible improvement in OS of 6.2 months as opposed to 5.3 months with supportive treatment alone [Citation16]. More recently, 5-FU/LV was examined in the second-line scenario with or without nal-irinotecan in the NIFTY phase IIb study. The revised extended follow-up data showed lesser differences, with the masked ICR-assessed mPFS reported as 4.2 and 1.7 months, respectively, and the BICR-assessed mPFS of 7.1 months with nal-irinotecan vs. 1.4 months without nal-irinotecan [Citation17].

A new stage II study assessing albumin-bound paclitaxel added to GEM-cisplatin in BTC detailed a medium overall survival of 19.2 months [Citation133] in the meanwhile a stage III trial with this triplet versus the normal GEM-cisplatin doublet is not ongoing (NCT03768414) [Citation134].

It is important to note that while these strategies show promise, overcoming chemoresistance remains a complex and ongoing research area. Clinical trials and translational studies are warranted to validate the effectiveness of these approaches in BTC and PCs.

3. Molecular mechanisms of resistance to target therapy

In the era of precision medicine, targeted therapies have emerged as a promising frontier in the treatment of various malignancies. These therapies, designed to specifically target aberrant molecular pathways driving cancer growth, have demonstrated remarkable efficacy in numerous tumors [Citation135,Citation136]. However, a formidable challenge persists: the development of resistance to these targeted interventions. As with conventional chemotherapies, cancer cells exhibit a remarkable capacity to adapt and evolve, rendering once-effective targeted therapies progressively less potent [Citation137]. Broadly, there are three main mechanisms underlying resistance to target therapies [Citation137] ().

  1. On-target alterations: these include changes in the target receptor that cause the drugs to not be able to inhibit the pathway. A striking example of this mechanism can be found in EGFR mutated non-small cell lung cancer (NSCLC): the use of adenosine triphosphate (ATP)-competitive inhibitors such as gefitinib and erlotinib is often associated with the development of a T790M point mutation in the EGFR gene, causing an increase in affinity for the physiologic substrate ATP and thus decreasing the inhibition on the downstream pathway by the target drugs [Citation138]. Other than single amino acid changes, on-target alteration can also include RNA alternative splicing, as has been seen in melanoma BRAF V600E patients treated with TKI, or gene amplification, as seen in imatinib-treated patients with chronic myeloid leukemia [Citation139,Citation140] ().

  2. Off-target alterations: to bypass a drug’s inhibition on a specific survival pathway, cancer cells often exhibit upregulation of different proliferation pathways. For example, BRAF V600E melanoma patients have to be treated with both BRAF and MEK inhibitors because the use of BRAF monotherapy leads to an upregulation in NRAS and a consequent mitogen-activated protein kinase (MAPK) reactivation [Citation141]. Similarly, NSCLC treated with EGFR, anaplastic lymphoma kinase (ALK), RET, or reactive oxygen species (ROS)-1 inhibitors, often display a secondary MET amplification driving resistance [Citation142] ().

  3. Phenotypic transformation: histological changes have been described in several cancers treated with TKI [Citation143]. EGFR mutated and ALK fusion-positive NSCLC treated with TKI can exhibit a transformation to small cell lung cancer, sometimes retaining the original mutation [Citation144]. More commonly the phenotypic transformation takes the shape of an EMT, in which alterations in gene expression and transcriptional mechanisms give rise to cells with mesenchymal properties such as changes in cells’ polarity, weakened cellular adhesion, increased migratory capacity, and cytoskeletal remodeling [Citation145] ().

To summarize, targeted therapies have managed to demonstrate remarkable efficacy by specifically attacking the aberrant molecular pathways that drive cancer growth. However, on-target, off-target and phenotypic changes can allow cancer cells to bypass the inhibition from target therapies. A comprehensive understanding of the underlying resistance mechanisms is essential to develop strategies to overcome them or to preemptively address them, thereby maximizing the long-term effectiveness of targeted therapies in the changing landscape of precision medicine.

3.1. Resistance to target therapy in biliary tract cancer

Although target therapies were added to the standard treatment paradigm of BTC only recently, the problem of treatment resistance has already emerged prominently. Due to a few clinical trials, research studies, and case reports focusing on different targets and drugs, we have garnered limited insight into the mechanisms that drive this phenomenon in BTC. On-target alterations have been detected using circulating tumor DNA (ctDNA) in liquid biopsy in patients treated with FGFR inhibitors. A small case series on eight patients with FGFR fusion or amplification reported that, at disease progression, 5/8 patients had developed mutations in the kinase domain of FGFR [Citation146]. Interestingly each patient had up to 9 mutations, indicating a wide heterogeneity of resistance mechanisms. A similar pattern of development of multiple point mutations per patient was observed in the ctDNA of 2 out of 3 FGFR2 fusion-positive CCA patients progressing after treatment with infigratinib; furthermore, autopsy data on one of the patients showed that all 12 sampled metastases retained the FGFR fusion alteration but with different interlesional mutations, pointing to the idea that progression was not driven by selection of FGFR fusion-negative clones but from different convergent resistance mutations [Citation147].

Regarding off-target alteration, it has been suggested that the co-presence of other mutations could be a cause for primary resistance to FGFR inhibitor, for example, 9 patients with both FGFR2 fusions and p53 mutations did not have an objective response to pemigatinib, and patients with CDKN2A or PBRM1 alteration had a shorter PFS [Citation148]. Consistent with findings on colon cancer, primary resistance to EGFR therapy has also been noticed in CCA patients harboring a K-RAS mutation [Citation149](). Furthermore, off-target secondary resistance to FGFR inhibitors has been observed in both preclinical models, with a reactivation of MEK/ERK [Citation150], and in the clinical setting, with the activation of the PI3K/AKT pathway [Citation151].

In BTC there are complex interlaying mechanisms between the tumor microenvironment and it has been observed that cancer-associated fibroblasts can favor epithelial–mesenchymal transition, resistance to chemotherapy and cancer progression [Citation152]. Furthermore, a preclinical paper suggests that epithelial–mesenchymal transition and acquisition of cancer stem cell properties can be a resistance mechanism in CCA cells treated with EGFR inhibitor erlotinib [Citation153].

3.2. Resistance to target therapy in pancreatic cancer

The genetic landscape of PC has been well analyzed in search of targetable mutations, but monotherapy treatment with target drugs did not have much success in PC, possibly due to many dysregulated pathways with complex and multiple cross talks and the fact that the most commonly mutated genes do not have a corresponding target drug [Citation58]. Thus, information on resistance mechanisms pertaining to PC specifically is scarce and limited to the few drugs that have been used in more advanced clinical trials.

For example, although KRAS alterations are present in around 90% of PC cases (), only a very small subset exhibits the G12C alteration that makes it susceptible to the target inhibitor sotorasib [Citation154]. However, even in these selected patients, the recorded ORR to second or further-line target therapy was only 21% [Citation61]. Preclinical data point to the activation of alternative signaling pathways and the development of new mutations in the KRAS gene as possible secondary resistance mechanisms to KRAs inhibitor, ongoing trials are evaluating allele-specific inhibitors and pan-(K)RAS inhibitors against the more common allele variants G12D, G12V, and G12R 290 [Citation155].

Furthermore, the presence of KRAS alteration has been linked to resistance to another type of target treatment, EGFR inhibitor. For example, the combination of anti-EGFR antibody nimotuzumab plus GEM had an OS advantage only in KRAS wild-type patients [Citation156]. Many other trials with EGFR inhibitors (both TKI and monoclonal antibody) failed to prove a survival benefit in unselected pancreatic patients, again, probably due to alternative active pathways [Citation157]. It should also be noted that, although the combination of erlotinib in association with GEM is associated with an OS gain [Citation28], this regimen is not often used in clinical practice as the benefit in the trial was minimal.

One of the few target therapies that has been approved in PC is the PARP inhibitor, olaparib, in the setting of BRCA1/2 germline-mutated patients that had not progressed after platinum-based first-line therapy. This is quite a rare scenario in which two mechanisms of DNA repair are blocked, one related to the BRCA gene and one to PARP enzymes, making the cancer cells extremely susceptible to DNA damage mechanisms. Despite this encouraging premise, maintenance therapy with olaparib after platinum-based first-line therapy was not associated with a gain in OS (18.9 vs 18.1 months), but only a PFS gain (7.4 vs 3.8 months in the placebo arm) [Citation27]. Although clinical data on PC patients are not available, resistance mechanisms to olaparib have been studied in pancreatic cell lines, pointing to the development of multiple genetic alterations including upregulation of MDR genes and phenotype changes such as EMT, furthermore, in other cancer, resistance to olaparib was also associated with 53BP1 downregulation and BRCA reversion mutations [Citation158].

3.3. Overcoming target therapy resistance

Understanding the molecular mechanisms of resistance to targeted therapy is crucial for developing strategies to overcome or prevent resistance (). The first issue that needs to be addressed is how to recognize which resistance mechanisms are at play in each patient. Given the wide spectrum of resistance mechanisms and the high intra-tumoral heterogeneity of both BTC and PC, it is unlikely that a single lesion biopsy will unravel the full complexity of the resistance mechanisms that can arise in each patient. Thus, it is crucial to implement tools that will give a more comprehensive picture of the resistance mechanisms during cancer progression. In this context, the implementation of liquid biopsy is emerging as a very valuable tool.

Table 4. Overcoming target therapy resistance in BTC and PC.

A study on 23 patients with BTC reported an overall blood/tissue concordance at diagnosis of 74%, ranging from 55% per ECC and 92% for ICC [Citation159], indicating the need for more in depth analysis on the applicability of liquid biopsy, especially in extrahepatic CCA. In PC, a meta-analysis on the use of liquid biopsy found an overall sensitivity of 70% and a specificity of 86% when compared to molecular analysis of tissue specimen; when looking at studies on KRAS mutations only, the sensitivity of liquid biopsy was a bit lower, around 65%, but with a high 91% specificity [Citation160]. Of note, 78% of mutations detected in circulating free DNA were not found in the lesion biopsy, again confirming the likely inadequacy of single lesion biopsy [Citation160].

Detecting the type or types of ongoing resistance mechanisms is only part of the problem, as then we need viable strategies to combat them ().

The development of target drugs that are active against the most common resistance mutations could be a noteworthy approach (). For example, in EGFR-mutated NSCLC osimertinib, an irreversible TKI, active against a common resistance mutation associated with previous inhibitors, has led to significant increase in PFS [Citation161]. In BTC this has been partially done with the development of futibatinib, an FGFR irreversible inhibitor active against some secondary kinase domain mutations arising from previous treatment with target therapy [Citation162]. Furthermore, drugs such as erdafitinib are being tested not only in FGFR2 fusion positive cases but also in FGFR1–4 alteration, and another FGFR inhibitor, tinengotinib, has recently proved to be useful also in patients pretreated with FGFR inhibitors [Citation163,Citation164]. However, we still do not have a complete picture of which mutations can arise and will likely respond to each FGFR inhibitor on the market, so we are still away from a truly personalized approach.

Multiple new drugs are currently being tested. For example, the novel KRAS inhibitor (MRTX1133), targeting the G12D mutation, has already demonstrated preclinical efficacy, showing tumor regression in the majority of tested cell line xenografts (8 out 11) and patient-derived xenografts [Citation165], and is now being tested in a clinical trials (NCT05737706) [Citation166]. Another pan-KRAS inhibitor called BI-3406, currently in phase-1 trials, disrupts the binding of KRAS to its activator SOS1 [Citation167], and similarly another KRAS G12X inhibitor called RMC-6236 is being tested in at least two phase 1 trials (NCT05379985, NCT06128551) [Citation168,Citation169]. Additionally, the SHP2 inhibitor TNO155 has shown promising safety and tolerability, and multiple clinical trials are underway to test its efficacy in combination with other therapies [Citation170,Citation171].

Another emerging therapy for PC involves targeting the Fanconi Anemia (FA) pathway, which is involved in DNA cross-link repair: ATR inhibitors, which inhibit the downstream signaling of the FA pathway, have been shown to be effective in preclinical studies when combined with chemotherapy agents such as GEM [Citation172,Citation173] ().

A different strategy consists in combining multiple target drugs to block multiple pathways, similar to what has already been done in other cancers like melanoma [Citation174] (). Such combination therapies can help overcome and prevent resistance by blocking alternative signaling pathways or compensatory mechanisms. In preclinical BTC cell models derived from patients harboring FGFR2 fusion and that had progressed on FGFR inhibitor monotherapy, there was a synergic activity of EGFR and FGFR inhibitors [Citation149]. Similarly, in a CCA cell line, a synergistic effect was observed with the use of a mTOR inhibitor plus a FGFR inhibitor [Citation151]. This cell line was derived from a patient progressing to FGFR monotherapy due to upregulation of the PI3K/AKT/mTOR pathway, which is frequently upregulated in FGFR constitutionally active cancer cells and that can act as a bypass mechanism when FGFR is inhibited [Citation151]. Again, this underlines the need to identify the mechanisms of resistance to allow for personalized treatments.

Similarly, in PC, the use of KRAS and MEK inhibitor has been associated with a compensatory activation of the AKT/mTOR pathway through cross talks proteins, thus the combination of either KRAS or MEK inhibitor along with an mTORC1/2 inhibitor has been used in preclinical models to prevent the activation of alternative pathways as a mechanism of resistance [Citation175]. Several trials investigating combination therapies are underway, mostly in basket trials on solid tumors including PC, and involving different targets such as SHP2, ERK, MEK, KRAS, EGFR, VEGF, and CD4/6 [Citation176–178] .Furthermore, given the use of olaparib in BRCA1/2 mutated patients, one study explored the use of mTOR inhibitors in combination with PARP inhibitors, pointing to a synergic effect of this combination even in BRCA wild-type cell lines [Citation179]. The combination of PARPi with immune checkpoint inhibitors has also been specifically used because BRCA insufficiency can trigger the production of type I interferon and pro-inflammatory cytokines, which can lead to an innate immune response that is dependent on the stimulator of interferon genes [Citation180]. Furthermore, in a dose-dependent manner, clinical models have shown that PARP inhibition up-regulates PD-L1 and inactivates glycogen synthase kinase-3 (GSK3). As a result, T-cell activation is suppressed, which increases the death of cancer cells [Citation181]. A phase 1/2 trial comparing PARPi niraparib plus nivolumab or ipilimumab as maintenance therapy after chemotherapy showed promising results in PFS for the niraparib–ipilimumab combination [Citation182] and more trials are ongoing with olaparib plus pembrolizumab (NCT05093231, NCT04548752, NCT04753879) [Citation183–185]

The combination of target therapy and immunotherapy is also being investigated (). Currently, a phase-2 study (NCT04753879) is actively enrolling participants to evaluate the effectiveness of maintenance olaparib in combination with pembrolizumab after low-dose GEM, nab-paclitaxel, capecitabine, cisplatin, and irinotecan in previously untreated metastatic PC patients [Citation185]. Additionally, the National Cancer Institute is conducting a phase-2 trial (NCT04548752) comparing the use of olaparib alone versus the combination of olaparib and pembrolizumab in patients with metastatic germline BRCA1/2-mutated PC. Another trial (NCT04493060) is currently investigating the use of niraparib and dostarlimab, an anti-PD-1 antibody, in metastatic PC patients with BRCA1/2 and PALB2 mutations [Citation186]. In BTC some ongoing trials include the anti IDH1 ivosidenib with nivolumab plus ipilimumab and the combination of PARP inhibitor olaparib plus durvalumab (NCT05921760, NCT03991832) [Citation187,Citation188]

In conclusion, resistance to targeted therapy is a complex phenomenon driven by various molecular mechanisms. By unraveling these mechanisms and developing innovative strategies, we can enhance the effectiveness of targeted therapies and improve patient outcomes.

4. Mechanisms of resistance to immunotherapy

Immunotherapy, heralded as a revolutionary paradigm in cancer treatment, has redefined the landscape of oncology by harnessing the intricate interplay between the immune system and malignant cells [Citation189]. By empowering the body’s immune defenses to recognize and combat cancer, immunotherapeutic agents have yielded unprecedented responses across a spectrum of malignancies. However, in PC, this class of drugs had to contend with high levels of primary resistance and the quick emergence of secondary resistance. The phenomenon of resistance to immunotherapy represents a complex interplay of biological mechanisms, whereby tumors employ a variety of strategies to evade or subvert the immune system’s potent antitumor effects. Understanding the diverse molecular and cellular mechanisms that confer resistance to immunotherapy is paramount for advancing the effectiveness of these groundbreaking treatments. Broadly speaking there are two main components associated with immune evasion [Citation190–192]. First, cancer cells possess intrinsic factors that play a crucial role in antigen presentation, a process vital for the immune system to recognize and eliminate cancer cells. However, loss of tumor neoantigen, changes in antigen presentation and processing, and hyperexpression of immune checkpoint proteins, can all help in the immune evasion. Furthermore, alteration of intracellular signaling due to genetic mutation, phenotypic transformation (such as EMT or a reversion to stem cell phenotype), and the secretion of metabolites to modify the tumor microenvironment are all possible causes of immune resistance as well. Second, the TME significantly shapes the presence, composition, and function of tumor infiltrating lymphocytes and other immune cells. The expression of immunosuppressive factors by the cancer cells, the presence of dysfunctional blood vessels, and a hostile, often hypoxic, environment are some of the factors contributing to immune evasion.

By illuminating the dynamic adaptations that underlie resistance, we aim to catalyze the development of strategies capable of overcoming these barriers and extending the transformative potential of immunotherapeutic approaches in the fight against BTC and PC.

4.1. Resistance to immunotherapy in BTC

Immunotherapy has been proven to prolong survival in BTC and has now become part of the first-line treatment, in combination with cisplatin and GEM. However, the number of patients responding to treatment is less than 30%, pointing to high levels of primary resistance to treatment (), and biomarkers of response that have been used in other cancers, such as PD-L1, have not been useful in BTC [Citation5,Citation7].

As BTCs are known to be very heterogeneous, several attempts have been made to classify them and possibly predict a response to therapy. For example, a genomic analysis divided BTC into 4 different categories, and it points out the presence of a cluster (cluster 4) as having both a high mutational load, causing elevated levels of neoantigens, and also having a high expression of immune checkpoint genes involved in suppressing an immune response [Citation193]. Furthermore, a paper on TME in iCCA points out the presence of four subtypes: the I1 immune-desert subtype is associated with feeble tumoral and stromal immune signaling, the I2 immunogenic subtype has a strong lymphoid and myeloid response, I3 myeloid subtypes displays only a strong myeloid response and the I4 mesenchymal subtypes is associated with activated fibroblasts, EMT, stem cells like features and neoangiogenesis174. This classification reflects survival rates with the I4 subtype having shorter survivals and I2 subtypes having better survival rates [Citation194]. We can then speculate that different subtypes could respond differently to immunotherapy, but no correlation between clinical data and response to different therapies is available.

Subclassification aside, several papers point to an intrinsic ability of BTC cells to suppress an immune response (). The secretion of PDGF-D and TGF-β by cancer cells causes the activation of CAFs, which in turn promotes the creation of a pro-inflammatory response with low CD8+ T cells and antigen presenting cells, and high Tregs and myeloid-derived suppressor cells [Citation195]. Furthermore, around half of BTC were found to have a low antigen presenting molecules (MCH)-1 expression, associated with worse survival rates and the levels of MCH-1 were also correlated with tumor associated lymphocytes and macrophages [Citation196] ().

Figure 4. Immunoresistance mechanism in BTC. tumor cells secrete PDGF and TGF-β which helps to recruit a large amount of TME CAFs through PDGFR-β signaling, these activated CAFs can release (1) proinflammatory compounds such as TGF-β and TNF-α that reinforce production by part of the tumor cells themselves; (2) ECM remodeling factors including MMP1, MMP2, MMP3, and MMP9; (3) growth factors including EGF, PDGF-B, and angiogenic factors, such as VEGF. VEGF isoforms, in turn, activate multiple VEGFRs (VEGFR1–3) that are found in a wide variety of endothelial and lymphatic cells, immune cells, and tumor cells themselves. Created with Biorender.com.

Figure 4. Immunoresistance mechanism in BTC. tumor cells secrete PDGF and TGF-β which helps to recruit a large amount of TME CAFs through PDGFR-β signaling, these activated CAFs can release (1) proinflammatory compounds such as TGF-β and TNF-α that reinforce production by part of the tumor cells themselves; (2) ECM remodeling factors including MMP1, MMP2, MMP3, and MMP9; (3) growth factors including EGF, PDGF-B, and angiogenic factors, such as VEGF. VEGF isoforms, in turn, activate multiple VEGFRs (VEGFR1–3) that are found in a wide variety of endothelial and lymphatic cells, immune cells, and tumor cells themselves. Created with Biorender.com.

It should also be noted that the presence of certain mutations could influence immune response. For example, IDH1 mutations cause an accumulation of the oncometabolite D-2-hydroxyglutarate, and, at least in gliomas, is associated with low CD8+ T cells and repression of the tumor immune system [Citation197]. Furthermore, FGFR signaling has been associated with a decrease in MCH-II, upregulation of PD-L1 and increase of regulatory T cells, again leading to an immune suppressive environment [Citation198].

To conclude, the causes of immune evasion in BTC are numerous (); however, data on how these mechanisms relate to response to immunotherapy is lacking.

4.2. Resistance to immunotherapy in pancreatic cancer

Immunotherapy is not effective for PC, despite the increased knowledge of the genomic landscape and of the complex TME (). Although it can be considered in patients with MSI or high tumor mutational burden, in this small subset of patients (around 2%), ORR to pembrolizumab has been reported between 62% and 18%, likely thanks to the high number of neoantigens being produced, capable of stimulating the immune system [Citation31,Citation199].

Figure 5. Immunoresistance mechanism in PC. (a) The tumor cells recruit and malignant the fibroblasts and macrophages of the tumor environment, thus the extracellular matrix becomes highly fibrotic and creates a physical barrier that prevents the infiltration of T cells into the tumor. (b) Immune evasion tactics in pancreatic cancer include: KRAS produced by the tumor cell reduces the expression of MHC I; just as miRNA-containing exosomes from tumor cells silence the expression of MHC II molecules in dendritic cells; and increased expression of antiphagocytic molecules such as CD47 prevents APC processing and tumor clearance. (c) The tumor suppresses immune responses through the expression of PD-L1, the recruitment of MDSC that activates regulatory T cells and inhibits T cells and NK cells, as well as the high expression of IDO that also allows them to block to the T cells. Created with Biorender.com.

Figure 5. Immunoresistance mechanism in PC. (a) The tumor cells recruit and malignant the fibroblasts and macrophages of the tumor environment, thus the extracellular matrix becomes highly fibrotic and creates a physical barrier that prevents the infiltration of T cells into the tumor. (b) Immune evasion tactics in pancreatic cancer include: KRAS produced by the tumor cell reduces the expression of MHC I; just as miRNA-containing exosomes from tumor cells silence the expression of MHC II molecules in dendritic cells; and increased expression of antiphagocytic molecules such as CD47 prevents APC processing and tumor clearance. (c) The tumor suppresses immune responses through the expression of PD-L1, the recruitment of MDSC that activates regulatory T cells and inhibits T cells and NK cells, as well as the high expression of IDO that also allows them to block to the T cells. Created with Biorender.com.

One culprit of immunotherapy’s discouraging results is certainly the TME: PCs have a dense and prominent stroma, rich with immunosuppressive cells such as regulatory T-cells (Treg), suppressive myeloid cells [Citation200], CAFs and TAMs, but lack of effector T cells [Citation201](). However, different compositions of TME have been described, with more mature-intermediate-immature characteristics, that correlate with prognosis, so it is possible that different subtypes may have different responses to immunotherapy [Citation202]. Furthermore, PC cells often exhibit low expression of tumor antigens, limiting the ability of the immune system to mount an effective response, and they can upregulate immunosuppressive proteins such as PD-L1 and Cytotoxic T-Lymphocyte Antigen 4 [Citation202] ().

4.3. Overcoming immunotherapy resistance

Overcoming immunotherapy resistance in cancer is a major focus of ongoing research and clinical trials and several strategies are being explored to enhance the effectiveness of immunotherapy.

First of all, a better understanding of which patients will benefit from immunotherapy will certainly be helpful, but unfortunately, markers used in other cancers, such as PD-L1, did not prove to be useful. For example, the use of pembrolizumab in pretreated BTC patients was associated with ORR between 6% and 13% with no discernible association with PD-L1 expression [Citation203]. On the other hand, the use of nivolumab in the same subset of patients saw similar ORR results (11%) and a significant correlation between PFS and PD-L1 expression, however, most notably, all responders to treatment had MSI [Citation204]. Currently, both in BTC and PC, the use of immunotherapy alone is only recommended in MSI cases, however, there could be other subsets of patients that could benefit from it. For example, a small case series on 12 BTC or PC patients with homologous recombination deficiency showed that the use of combination immunotherapy with nivolumab and ipilimumab was associated with an ORR of 42%, with 4 patients reaching a complete response. Furthermore, the responders had a much higher level of TIL compared to non-responders, with a T cell inflamed signature on RNA expression analysis [Citation205]. Furthermore, as stated before, several papers have tried to subclassify BTC and PC according to the different levels of immune suppression, but so far these classifications were not correlated to response to immunotherapy [Citation193,Citation194,Citation202] ().

Table 5. Overcoming immunoresistance in BTC and PC.

Other than a better selection of patients, equally important is the development of better and more effective approaches to stimulate the immune system and combat immune resistance.

Over the years, a viable strategy has been the use of combination therapies (). The combination of different drugs that can target multiple immune checkpoints has had conflicting results: for example, the use of anti PD-L1 and anti CTLA4 was associated with ORR of around 20% when involving nivolumab and ipilimumab [Citation206], while with the combination of durvalumab and tremelimumab the ORR was less than 5% [Citation207]. Moreover, other combinations are being tested with drugs working on different mechanisms of action, such as immunotherapy plus targeted therapy or chemotherapy. As stated before, the combination of chemo-immunotherapy is now the standard first-line in BTC, but many more are being evaluated such as anti VEGFR regorafenib + avelumab, or pembrolizumab + Lenvatinib, or chemotherapy plus lenvatinib and anti-PD-1 toripalimab [Citation208–210]. Furthermore, given the immunosuppressive nature of some genetic alterations, the idea of combining target inhibitor and immunotherapy is being explored: anti IDH1 Ivosidenib + nivolumab, anti FGFR2 futibatinib + pembrolizumab, anti MEK cobimetinib + atezolizumab, or the implementation of chimeric antigen receptor-engineered autologous T (CAR-T) cell immunotherapy directed against EGFR positive cells, just cite a few [Citation195,Citation211] (). Furthermore, other than PD-L1 or CTLA4 inhibition, novel immune targets are being explored, alone or in combination with other immune-stimulating drugs, with the intent to modify the immunosuppressive effect of the tumor microenvironment and boost the effect of immune cells. Some of these targets are, for example: TIM-3, LAG-3, TIGIT, and VISTA plus in PC specifically CD40, CD11b, OX40, IDO, and B7/H3 [Citation212–215].

Many other innovative immune strategies are in the early stages of testing. For example, the use of vaccines or oncolytic viruses, to enhance antigen presentation and improve the recognition of tumor cells by the immune system [Citation216]. Furthermore, the use of adoptive cell transfer therapy is being explored in different types of solid tumors, including BTC and PC [Citation217]. It involves genetically modifying a patient’s own immune cells, such as T cells or natural killer cells, to express chimeric antigen receptors or T cell receptors that specifically recognize tumor antigens; These modified cells are then infused back into the patient to target and kill cancer cells. For example, an early trial with the combination of allogeneic natural killer cells in combination with pembrolizumab showed good tolerability and some antitumor activity [Citation218]. Finally, since epigenetic modifications can influence the expression of genes involved in immune response regulation, the modulation of these epigenetic changes through specific drugs has the potential to sensitize the tumor to immunotherapy and overcome resistance: for this reason, several new drugs such as DNA methyltransferase inhibitors, HDAi, BET inhibitors, and EZH2 inhibitors are being tested [Citation219] ().

Furthermore, several phase II studies are currently underway investigating the possibility of incorporating immunotherapy into the adjuvant treatment of BTC, as It is possible that an early use of immunotherapy could lead to less treatment resistance. Combinations being explored include capecitabine plus lenvatinib and tislelizumab (NCT05254847 [Citation220]), capecitabine plus camrelizumab and radiotherapy (NCT04333927 [Citation221]), and capecitabine with durvalumab and tremelimumab (NCT05239169 [Citation222]). Furthermore, a phase II trial (NCT04506281 [Citation223]) is ongoing using the combination of oripalimab plus gemcitabine-oxaliplatin plus lenvatinib, which was previously studied in advanced/metastatic cases as a treatment for resectable iCCA, with promising results [Citation210].

To conclude, the road to overcome resistance to immunotherapy is still long. The lack of predictive tools to implement a better selection of patients that will likely benefit from immunotherapy and the complex immunosuppressive environment of both BTC and PC, are significant challenges that need to be overcome. Hopefully, an even better understanding of the resistance mechanisms at play and the ever-changing new ideas and technologies being studied will allow for a breakthrough soon.

5. Conclusions

Treatment resistance in pancreatic and biliary tract cancer is a significant challenge in the field of oncology, as it concerns all available therapies, from chemotherapy, to target therapy and immunotherapy. As these cancers have limited treatment options, shedding light on these hidden mechanisms of treatment resistance can pave the way for groundbreaking advancements in the fight against cancer.

The molecular mechanisms of resistance are numerous, complex, and often intertwined, as they include processes that are inherently connected: genetic alterations, altered drug metabolism and efflux, changes in the tumor microenvironment, and phenotypic transformations (). Unraveling the complexities of these mechanisms is essential to develop strategies to overcome or prevent resistance, ushering in a new era of personalized medicine, where tailored treatments can effectively combat chemoresistant cancers.

Despite the existence of multiple current therapies that are being employed to combat different levels of chemoresistance (), it is imperative to emphasize the role of collaborative interdisciplinary research in the pursuit of overcoming treatment resistance in PC and BTC. Integrating insights from diverse scientific disciplines, including genomics, pharmacology, immunology, and computational biology, can provide a more comprehensive understanding of the intricate network of factors contributing to resistance. Furthermore, advancements in diagnostic tools and technologies, such as liquid biopsies and high-throughput sequencing, hold promise in identifying early markers of treatment resistance. These tools not only enhance our ability to monitor the dynamic changes in the molecular landscape of tumors but also enable timely adjustments to therapeutic interventions.

Combination therapies, modulation of the tumor microenvironment, new drugs and personalized medicine approaches are being explored as potential solutions (). To confirm the efficacy of these strategies, particularly in complex and heterogeneous cancers like BTC and PC, additional studies and clinical trials are required.

In conclusion, the fight against treatment resistance in PC and BTC necessitates a multi-faceted approach that encompasses not only therapeutic strategies but also collaborative research endeavors and the integration of cutting-edge technologies. By embracing the complexity of these challenges and leveraging the collective expertise of the scientific community, we can pave the way for transformative breakthroughs in the battle against chemoresistant cancers.

6. Expert opinion

Considering previous studies on drug resistance in pancreatic and biliary tract cancer, the scientific community is still far away from having a concise picture of how cancer cells are able to survive the treatments that are routinely administered to patients.

In this review, we provide a concise picture of the mechanisms that have been discovered so far, in order to underline the astounding number of mechanisms that have been observed and studied in these cancers. What is truly lacking then is a solid theory of how all these different mechanisms combine to make both pancreatic and biliary tract cancers so challenging to treat. Unfortunately, the development of a unified theory on resistance to therapy is made even more challenging by the high heterogeneity of these cancers, so that the main mechanisms of resistance can be different not only between patients but also within different metastatic lesions of the same patients, or even different tumor areas of a single nodule [Citation224,Citation225].

Given how complicated the topic is, perhaps a different approach needs to be taken to the issue. One suggestion is to address each patient as an individual rather than concentrating on PC or BTC in general. This translates into the notion of creating a tool that can assist in identifying the primary mechanisms at work to better target the cancer in practice. This might take the form of a liquid biopsy, but it might also be a deeper single-cell transcriptome study of the tissue sample or perhaps another as-yet-undeveloped technology [Citation160,Citation226]. Indeed, an article on identifying resistance mechanisms in gastrointestinal cancer points to a better clinical relevance of the results obtained with liquid biopsy compared to tissue biopsy, as a liquid biopsy gave a more complete picture of the various ongoing resistance mechanisms while tissue biopsy could only identify the mechanisms relevant to the small acquired tissue sample [Citation227]. Furthermore, to better probe intra-tumoral heterogeneity, analysis of transcriptomic data from single cells are being performed to predict drug sensitivity and test selective drugs that could overcome treatment resistance, although these types of tests are only recently starting to be implemented in the clinical setting [Citation228,Citation229].

Given the rapid advances in computer technology, another emerging possibility is the use of artificial intelligence (AI) to integrate all the data available on different treatment resistance mechanisms. The use of AI could enable us to truly have a unified picture of the mechanisms of resistance to therapy, and consequently, it could help point scientists in the right direction for the development of more useful strategies to overcome resistance [Citation230–232]. AI has already been used to identify genes that are likely to be involved in modifications related to drug resistance, including epigenetic changes, and to predict which drugs are more likely to advance from the preclinical to the clinical setting, helping reduce the costs and time spent on dead-end projects [Citation230,Citation231]. In the setting of PC, advances in AI have already been made regarding early diagnosis, both with biomarkers and radiological imaging, while in the early stages of both PC and BTC, AI is being used to aid surgeons in cases of uncertain diagnostic imaging or for planning of complex surgery [Citation233,Citation234]. Hopefully, the implementation of AI in both PC and BTC to better understand the mechanisms of drug resistance will be part of the near future.

A more in-depth knowledge of the resistance mechanisms is helpful to guide treatment decisions, but it is only useful when combined with the development of new drugs or improvement of drugs that are already available. Small steps in this direction have already been made, for example, the development of new generation FGFR inhibitors in BTC, active against some resistance mutation to previous drugs, or the use of maintenance therapy with PARP inhibitor olaparib in BRCA1/2 mutated PC patients [Citation235]. Although these therapies are only applicable to a small subset of patients, we believe that they should be seen as a success and as herald drugs in the new landscape of personalized medicine. Hopefully, this type of personalized treatment can continue to be explored, even though the challenges are not only scientific in nature but also economical, as the road to developing drugs that can only be used in a small subset of patients is often arduous.

To conclude, the current therapeutic options for PC and BTC, including chemotherapy, targeted therapy, and immunotherapy, have shown limited success in overcoming treatment resistance. Our expert opinion is that a deeper understanding of each patient’s resistance mechanisms could really help in implementing the right drugs to prolong survival. To make these tailored treatments possible, it is also paramount that scientists keep working on novel approaches. We believe that an improvement in treatment results for these aggressive malignancies is feasible through a deeper comprehension of the molecular causes of resistance, the creation of customized methods, and the investigation of innovative combination medicines.

Article highlights

  • Therapeutic options for BTC and PC include chemotherapy, targeted therapy, and immunotherapy, however the prognosis of these patients remains poor.

  • Resistance to these treatments is common and it may arise from diverse molecular mechanisms, such as genetic alterations, epigenetic modifications, altered drug metabolism and efflux, and changes in the tumor microenvironment. Identifying and overcoming these mechanisms is a major focus of research.

  • Addressing each patient individually, utilizing tools such as liquid biopsy or single-cell transcriptome studies can help identify primary mechanisms of resistance, allowing for targeted treatments.

  • The article explores the emerging possibility of using artificial intelligence to integrate data on different treatment resistance mechanisms, aiming for a unified understanding and guiding the development of more effective strategies.

  • Examples of success in personalized medicine, such as the development of new generation FGFR inhibitors in BTC and maintenance therapy with PARP inhibitor olaparib in BRCA1/2 mutated PC patients, are acknowledged.

Abbreviations

5-AZA=

5-Azacytidine

ABC=

ATP-binding Cassette

ACT=

Adoptive Cell Transfer

AI=

Artificial Intelligence

AKT=

Serine/Threonine Kinase

ALK=

Anaplastic Lymphoma Kinase

ATM=

Ataxia Telangiectasia Mutated

ATP=

Adenosine Triphosphate

Bcl-2=

B-Cell Lymphoma 2

BRAF=

B-Raf Proto-oncogene, Serine/Threonine Kinase

BTC=

Biliary Tract Cancer

CAFs=

Cancer-associated Fibroblasts

CCA=

Cholangiocarcinoma

ctDNA=

Circulating Tumor DNA

DCR=

Disease Control Rate

DNA=

Deoxyribonucleic Acid

DSB=

Double Strand Breaks

ECC=

Extrahepatic Cholangiocarcinoma

ECM=

Extracellular Matrix

EGFR=

Epidermal Growth Factor Receptor

EMT=

Epithelial-mesenchymal Transition

ERK=

Extracellular Regulated Kinase

FA=

Fanconi Anemia

FDA=

Food and Drug Administration

FGFR=

Fibroblast Growth Factor Receptor

GEM=

Gemcitabine

HA=

Hyaluronic Acid

hENT=

Human Equilibrative Nucleoside Transporter

HDR=

Homology‐directed Repair

ICC=

Intrahepatic Cholangiocarcinoma

IDH1=

Isocitrate Dehydrogenase 1

KRAS=

Kirsten Rat Sarcoma Virus

MAPK=

Mitogen-activated Protein Kinase

MCH=

Antigen Presenting Molecules

MSI=

Microsatellite Instability

NF-kβ=

Nuclear Factor Kappa-light-chain-enhancer of Activated B Cells

NSCLC=

Non-small Cell Lung Cancer

NTRK=

Neurotrophic Tyrosine Receptor Kinase

ORR=

Overall Response Rate

OS=

Overall Survival

PARP=

Poly (ADP-ribose) Polymerase

PARPi=

Poly (ADP-ribose) Polymerase Inhibitor

PC=

Pancreatic Cancer

PD-L1=

Programmed Death-ligand 1

PDX=

Patient-Derived Xenograft

PFS=

Progression Free Survival

PI3K=

Phosphoinositide-3-kinase

PLOD2=

2-Oxoglutarate 5-Dioxygenase 2

RNA=

Ribonucleic Acid

ROS=

Reactive Oxygen Species

Shh=

Sonic hedgehog

TAM=

Tumor Associated Macrophage

TGF-β=

Transforming Growth Factor Beta

TKI=

Tyrosine Kinase Inhibitor

TME=

Tumor Microenvironment

Treg=

Regulatory T-cells

VEGF=

Vascular Endothelial Growth Factor

ZEB1=

Zinc Finger E-box Binding Homeobox 1

Declaration of interest

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Author contributions

B Toledo and C Deiana wrote the manuscript. E Giovannetti revised and corrected thoroughly the manuscript. All authors revised the manuscript critically and agreed to the published version of the manuscript.

Acknowledgments

The images were made using Biorender.com.

Additional information

Funding

This paper is based upon work from COST Action, Identification of biological markers for prevention and translational medicine in pancreatic cancer (TRANSPAN), CA21116, supported by COST (European Cooperation in Science and Technology) (to B Toledo). B Toledo was supported by an Award of EMBO Scientific Exchange Grant ref.: 10383, 2023. This paper has been funded by Consejería de Economía, Conocimiento, Empresas y Universidad de la Junta de Andalucía and European Regional Development Fund (ERDF), ref. P18-FR-2470, from the Ministry of Science, Innovation and Universities [ref. RTI 2018-101309-B-C22], and from the Chair “Doctors Galera-Requena in cancer stem cell research” [CMC-CTS963]. This paper was partly funded by KWF Dutch Cancer Society, Associazione Italiana per la Ricerca sul Cancro AIRC, and by the Bennink Foundation (to E Giovannetti).

References

  • Banales JM, Marin JJG, Lamarca A, et al. Cholangiocarcinoma 2020: the next horizon in mechanisms and management. Nat Rev Gastroenterol Hepatol. 2020;17:557–588. doi: 10.1038/s41575-020-0310-z
  • Surveillance, Epidemiology, and End Results (SEER), Program populations (1969-2020) National Cancer Institute, DCCPS, Surveillance Research program. Cancer of the liver and intrahepatic bile duct - Cancer stat facts [internet]. SEER. 2022 [cited 2023 Oct 30]. Available from: https://seer.cancer.gov/statfacts/html/livibd.html
  • Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424. doi: 10.3322/caac.21492
  • Rawla P, Sunkara T, Gaduputi V. Epidemiology of pancreatic cancer: global trends, etiology and risk factors. World J Oncol. 2019;10:10–27. doi: 10.14740/wjon1166
  • Oh D-Y, Ruth HA, Qin S, et al. Durvalumab plus Gemcitabine and cisplatin in advanced biliary tract cancer. NEJM Evid. 2022;1:EVIDoa2200015.
  • Oh D-Y, He AR, Qin S, et al. 78P updated overall survival (OS) from the phase III TOPAZ-1 study of durvalumab (D) or placebo (PBO) plus gemcitabine and cisplatin (+ GC) in patients (pts) with advanced biliary tract cancer (BTC). Ann Oncol. 2022;33:S1462–S1463. doi: 10.1016/j.annonc.2022.10.114
  • Kelley RK, Ueno M, Yoo C, et al. Pembrolizumab in combination with gemcitabine and cisplatin compared with gemcitabine and cisplatin alone for patients with advanced biliary tract cancer (KEYNOTE-966): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2023;401:1853–1865. doi: 10.1016/S0140-6736(23)00727-4
  • Abou-Alfa GK, Sahai V, Hollebecque A, et al. Pemigatinib for previously treated, locally advanced or metastatic cholangiocarcinoma: a multicentre, open-label, phase 2 study. Lancet Oncol. 2020;21:671–684.
  • Vogel A, Sahai V, Hollebecque A, et al. O-2 pemigatinib for previously treated locally advanced or metastatic cholangiocarcinoma: final results from FIGHT-202. Ann Oncol. 2022;33:S379. doi: 10.1016/j.annonc.2022.04.443
  • Goyal L, Meric-Bernstam F, Hollebecque A, et al. Futibatinib for FGFR2-rearranged intrahepatic cholangiocarcinoma. N Engl J Med. 2023;388:228–239. doi: 10.1056/NEJMoa2206834
  • Javle M, Roychowdhury S, Kelley RK, et al. Infigratinib (BGJ398) in previously treated patients with advanced or metastatic cholangiocarcinoma with FGFR2 fusions or rearrangements: mature results from a multicentre, open-label, single-arm, phase 2 study. Lancet Gastroenterol Hepatol. 2021;6:803–815. doi: 10.1016/S2468-1253(21)00196-5
  • Abou-Alfa GK, Macarulla T, Javle MM, et al. Ivosidenib in IDH1-mutant, chemotherapy-refractory cholangiocarcinoma (ClarIdhy): a multicentre, randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol. 2020;21:796–807. doi: 10.1016/S1470-2045(20)30157-1
  • Harding JJ, Fan J, Oh D-Y, et al. Zanidatamab for HER2-amplified, unresectable, locally advanced or metastatic biliary tract cancer (HERIZON-BTC-01): a multicentre, single-arm, phase 2b study. Lancet Oncol. 2023;24:772–782. doi: 10.1016/S1470-2045(23)00242-5
  • Knox JJ, Lee CL, O’Kane G Targets and resistance mechanisms in biliary tract cancers. ASCO Dly News [Internet]. [cited 2023 Oct 5]. Available from: https://dailynews.ascopubs.org/do/10.1200/ADN.23.201274/full
  • Cho SM, Esmail A, Raza A, et al. Timeline of FDA-Approved targeted therapy for Cholangiocarcinoma. Cancers (Basel). 2022;14:2641. doi: 10.3390/cancers14112641
  • Lamarca A, Palmer DH, Wasan HS, et al. Second-line FOLFOX chemotherapy versus active symptom control for advanced biliary tract cancer (ABC-06): a phase 3, open-label, randomised, controlled trial. Lancet Oncol. 2021;22:690–701. doi: 10.1016/S1470-2045(21)00027-9
  • Yoo C, Kim K, Jeong JH, et al. Liposomal irinotecan plus fluorouracil and leucovorin versus fluorouracil and leucovorin for metastatic biliary tract cancer after progression on gemcitabine plus cisplatin (NIFTY): a multicentre, open-label, randomised, phase 2b study. Lancet Oncol. 2021;22:1560–1572. doi: 10.1016/S1470-2045(21)00486-1
  • Vogel A, Wenzel P, Folprecht G, et al. 53MO nal-IRI and 5-FU/LV compared to 5-FU/LV in patients with cholangio- and gallbladder carcinoma previously treated with gemcitabine-based therapies (NALIRICC – AIO-HEP-0116). Ann Oncol. 2022;33:S563–S564. doi: 10.1016/j.annonc.2022.07.081
  • Ying J, Chen J. Combination versus mono-therapy as salvage treatment for advanced biliary tract cancer: a comprehensive meta-analysis of published data. Crit Rev Oncol Hematol. 2019;139:134–142. doi: 10.1016/j.critrevonc.2019.01.001
  • Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817–1825.
  • Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-Paclitaxel plus Gemcitabine. N Engl J Med. 2013;369:1691–1703.
  • Wainberg ZA, Melisi D, Macarulla T, et al. NALIRIFOX versus nab-paclitaxel and gemcitabine in treatment-naive patients with metastatic pancreatic ductal adenocarcinoma (NAPOLI 3): a randomised, open-label, phase 3 trial. Lancet. 2023 [cited 2023 Oct 5]. Available from: https://www.thelancet.com/journals/lancet/article/PIIS0140-6736(23)01366-1/fulltext
  • Portal A, Pernot S, Tougeron D, et al. Nab-paclitaxel plus gemcitabine for metastatic pancreatic adenocarcinoma after Folfirinox failure: an AGEO prospective multicentre cohort. Br J Cancer. 2015;113:989–995. doi: 10.1038/bjc.2015.328
  • Chung MJ, Kang H, Kim HG, et al. Multicenter phase II trial of modified FOLFIRINOX in gemcitabine-refractory pancreatic cancer. World J Gastrointest Oncol. 2018;10:505–515. doi: 10.4251/wjgo.v10.i12.505
  • Gill S, Ko Y-J, Cripps C, et al. PANCREOX: a randomized phase III study of Fluorouracil/Leucovorin with or without oxaliplatin for second-line advanced pancreatic cancer in patients who have received gemcitabine-based chemotherapy. J Clin Oncol. 2016;34:3914–3920. doi: 10.1200/JCO.2016.68.5776
  • Wang-Gillam A, Li C-P, Bodoky G, et al. Nanoliposomal irinotecan with fluorouracil and folinic acid in metastatic pancreatic cancer after previous gemcitabine-based therapy (NAPOLI-1): a global, randomised, open-label, phase 3 trial. Lancet. 2016;387:545–557. doi: 10.1016/S0140-6736(15)00986-1
  • Golan T, Hammel P, Reni M, et al. Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. N Engl J Med. 2019;381:317–327. doi: 10.1056/NEJMoa1903387
  • Moore MJ, Goldstein D, Hamm J, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25:1960–1966. doi: 10.1200/JCO.2006.07.9525
  • Doebele RC, Drilon A, Paz-Ares L, et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1–2 trials. Lancet Oncol. 2020;21:271–282. doi: 10.1016/S1470-2045(19)30691-6
  • Hong DS, DuBois SG, Kummar S, et al. Larotrectinib in patients with TRK fusion-positive solid tumours: a pooled analysis of three phase 1/2 clinical trials. Lancet Oncol. 2020;21:531–540. doi: 10.1016/S1470-2045(19)30856-3
  • Marabelle A, Le DT, Ascierto PA, 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;38:1–10. doi: 10.1200/JCO.19.02105
  • Leroux C, Konstantinidou G. Targeted therapies for pancreatic cancer: overview of current treatments and new opportunities for personalized oncology. Cancers (Basel). 2021;13:799. doi: 10.3390/cancers13040799
  • Zheng H-C. The molecular mechanisms of chemoresistance in cancers. Oncotarget. 2017;8:59950–59964. doi: 10.18632/oncotarget.19048
  • Wu C-E, Pan Y-R, Yeh C-N, et al. Targeting P53 as a future strategy to overcome gemcitabine resistance in biliary tract cancers. Biomolecules. 2020;10:1474. doi: 10.3390/biom10111474
  • Sanchon-Sanchez P, Briz O, Macias RIR, et al. Evaluation of potential targets to enhance the sensitivity of cholangiocarcinoma cells to anticancer drugs. Biomed Pharmacother Biomed Pharmacother. 2023;168:115658. doi: 10.1016/j.biopha.2023.115658
  • Zeekpudsa P, Kukongviriyapan V, Senggunprai L, et al. Suppression of NAD(P)H-quinone oxidoreductase 1 enhanced the susceptibility of cholangiocarcinoma cells to chemotherapeutic agents. J Exp Clin Cancer Res CR. 2014;33:11. doi: 10.1186/1756-9966-33-11
  • Tannapfel A, Sommerer F, Benicke M, et al. Mutations of the BRAF gene in cholangiocarcinoma but not in hepatocellular carcinoma. Gut. 2003;52:706–712. doi: 10.1136/gut.52.5.706
  • Peng J, Fang S, Li M, et al. Genetic alterations of KRAS and TP53 in intrahepatic cholangiocarcinoma associated with poor prognosis. Open Life Sci [Internet]. 2023;18 [cited 2023 Nov 15]. Available from: https://www.degruyter.com/document/doi/10.1515/biol-2022-0652/html
  • Buckarma EH, Werneburg NW, Conboy CB, et al. The YAP-interacting phosphatase SHP2 can regulate transcriptional coactivity and modulate sensitivity to chemotherapy in cholangiocarcinoma. Mol Cancer Res MCR. 2020;18:1574–1588. doi: 10.1158/1541-7786.MCR-20-0165
  • Abou-Alfa GK, Bibeau K, Schultz N, et al. Effect of FGFR2 alterations on overall and progression-free survival in patients receiving systemic therapy for Intrahepatic Cholangiocarcinoma. Target Oncol [Internet]. 2022 [cited 2022 Sep 21]; Available from];17(5):517–527. doi: 10.1007/s11523-022-00906-w
  • Brandi G, Deiana C, Galvani L, et al. Are FGFR and IDH1-2 alterations a positive prognostic factor in intrahepatic cholangiocarcinoma? An unresolved issue. Front Oncol. 2023;13:1137510. doi: 10.3389/fonc.2023.1137510
  • Kim H, Kim JY, Park KU. Clinical implications of BRCA mutations in advanced biliary tract cancer. Oncology. 2023;101:41–48. doi: 10.1159/000527525
  • Mao Y, Huang X, Shuang Z, et al. PARP inhibitor olaparib sensitizes cholangiocarcinoma cells to radiation. Cancer Med. 2018;7(4):1285–1296. doi: 10.1002/cam4.1318
  • Banales JM, Cardinale V, Carpino G, et al. Cholangiocarcinoma: current knowledge and future perspectives consensus statement from the European Network for the Study of Cholangiocarcinoma (ENS-CCA). Nat Rev Gastroenterol Hepatol. 2016;13:261–280. doi: 10.1038/nrgastro.2016.51
  • Yu Y, Zhang M, Wang N, et al. Epigenetic silencing of tumor suppressor gene CDKN1A by oncogenic long non-coding RNA SNHG1 in cholangiocarcinoma. Cell Death Dis. 2018;9:746. doi: 10.1038/s41419-018-0768-6
  • Sittithumcharee G, Suppramote O, Vaeteewoottacharn K, et al. Dependency of cholangiocarcinoma on cyclin D-Dependent kinase activity. Hepatology. 2019;70:1614–1630. doi: 10.1002/hep.30704
  • Puik JR, Meijer LL, Le Large TY, et al. miRNA profiling for diagnosis, prognosis and stratification of cancer treatment in cholangiocarcinoma. Pharmacogenomics. 2017;18:1343–1358. doi: 10.2217/pgs-2017-0010
  • Meng F, Henson R, Lang M, et al. Involvement of human micro-RNA in growth and response to chemotherapy in human cholangiocarcinoma cell lines. Gastroenterology. 2006;130:2113–2129. doi: 10.1053/j.gastro.2006.02.057
  • Toyota Y, Iwama H, Kato K, et al. Mechanism of gemcitabine-induced suppression of human cholangiocellular carcinoma cell growth. Int J Oncol. 2015;47:1293–1302. doi: 10.3892/ijo.2015.3118
  • Carotenuto P, Hedayat S, Fassan M, et al. Modulation of biliary cancer chemo-resistance through MicroRNA-mediated rewiring of the expansion of CD133+ cells. Hepatology. 2020;72:982–996. doi: 10.1002/hep.31094
  • Toledo B, Picon-Ruiz M, Marchal JA, et al. Dual role of fibroblasts educated by tumour in cancer behavior and therapeutic perspectives. Int J Mol Sci. 2022;23:15576.
  • Fabris L, Sato K, Alpini G, et al. The tumor microenvironment in cholangiocarcinoma progression. Hepatology. 2021;73:75–85. doi: 10.1002/hep.31410
  • Obata T, Tsutsumi K, Ueta E, et al. MicroRNA-451a inhibits gemcitabine-refractory biliary tract cancer progression by suppressing the MIF-mediated PI3K/AKT pathway. Mol Ther Nucleic Acids. 2023;34:102054. doi: 10.1016/j.omtn.2023.102054
  • Yang C, Xu M, Shen H-J, et al. Potential biomarkers for sensitivity of gallbladder cancer cells to gemcitabine. Int J Clin Exp Pathol. 2014;7:521–528.
  • Okumura Y, Noda T, Eguchi H, et al. Hypoxia-induced PLOD2 is a key regulator in Epithelial-Mesenchymal transition and chemoresistance in biliary tract cancer. Ann Surg Oncol. 2018;25:3728–3737. doi: 10.1245/s10434-018-6670-8
  • Wang S, Zheng Y, Yang F, et al. The molecular biology of pancreatic adenocarcinoma: translational challenges and clinical perspectives. Signal Transduct Target Ther. 2021;6:1–23. doi: 10.1038/s41392-021-00659-4
  • Halbrook CJ, Lyssiotis CA, Pasca di Magliano M, et al. Pancreatic cancer: advances and challenges. Cell. 2023;186:1729–1754. doi: 10.1016/j.cell.2023.02.014
  • Garrido-Laguna I, Hidalgo M. Pancreatic cancer: from state-of-the-art treatments to promising novel therapies. Nat Rev Clin Oncol. 2015;12:319–334. doi: 10.1038/nrclinonc.2015.53
  • Fedele C, Ran H, Diskin B, et al. SHP2 inhibition prevents adaptive resistance to MEK inhibitors in multiple cancer models. Cancer Discov. 2018;8:1237–1249. doi: 10.1158/2159-8290.CD-18-0444
  • Ardalan B, Azqueta JI, England J, et al. Potential benefit of treatment with MEK inhibitors and chemotherapy in BRAF-mutated KRAS wild-type pancreatic ductal adenocarcinoma patients: a case report. Mol Case Stud. 2021;7:a006108. doi: 10.1101/mcs.a006108
  • Strickler JH, Satake H, George TJ, et al. Sotorasib in KRAS p.G12C–mutated advanced pancreatic cancer. N Engl J Med. 2023;388:33–43. doi: 10.1056/NEJMoa2208470
  • Pan M, Jiang C, Zhang Z, et al. TP53 gain-of-function and non–gain-of-Function mutations are associated with differential prognosis in advanced pancreatic ductal adenocarcinoma. JCO Precis Oncol. 2023;e2200570. doi: 10.1200/PO.22.00570
  • Rosen MN, Goodwin RA, Vickers MM. BRCA mutated pancreatic cancer: a change is coming. World J Gastroenterol. 2021;27:1943–1958. doi: 10.3748/wjg.v27.i17.1943
  • Wong W, Raufi AG, Safyan RA, et al. BRCA mutations in pancreas cancer: spectrum, current management, challenges and future prospects. Cancer Manag Res. 2020;12:2731–2742. doi: 10.2147/CMAR.S211151
  • Chen C-C, Kass EM, Yen W-F, et al. ATM loss leads to synthetic lethality in BRCA1 BRCT mutant mice associated with exacerbated defects in homology-directed repair. Proc Natl Acad Sci, USA. 2017;114(29):7665–7670. doi: 10.1073/pnas.1706392114
  • Shoucair S, Baker AR, Yu J. Germline variants in DNA damage repair genes: an emerging role in the era of precision medicine in pancreatic adenocarcinoma. Ann Gastroenterol Surg. 2022;6(1):7–16. doi: 10.1002/ags3.12514
  • Blair AB, Groot VP, Gemenetzis G, et al. BRCA1/BRCA2Germline mutation carriers and sporadic pancreatic ductal adenocarcinoma. J Am Coll Surg. 2018;226(4):630. doi: 10.1016/j.jamcollsurg.2017.12.021
  • Park W, Chen J, Chou JF, et al. Genomic methods identify homologous recombination deficiency in pancreas adenocarcinoma and optimize treatment selection. Clin Cancer Res. 2020;26(13):3239–3247. doi: 10.1158/1078-0432.CCR-20-0418
  • O’Reilly EM, Lee JW, Zalupski M, et al. Randomized, multicenter, phase II trial of gemcitabine and cisplatin with or without veliparib in patients with pancreas adenocarcinoma and a Germline BRCA/PALB2 Mutation. J Clin Oncol. 2020;38:1378–1388. doi: 10.1200/JCO.19.02931
  • Baylin SB, Jones PA. Epigenetic determinants of cancer. Cold Spring Harb Perspect Biol. 2016;8:a019505. doi: 10.1101/cshperspect.a019505
  • Wang SS, Xu J, Ji KY, et al. Epigenetic alterations in pancreatic cancer metastasis. Biomolecules. 2021;11:1082. doi: 10.3390/biom11081082
  • Kurdistani SK, Tavazoie S, Grunstein M. Mapping global histone acetylation patterns to gene expression. Cell. 2004;117:721–733. doi: 10.1016/j.cell.2004.05.023
  • Borchert GM, Lanier W, Davidson BL. RNA polymerase III transcribes human microRnas. Nat Struct Mol Biol. 2006;13:1097–1101. doi: 10.1038/nsmb1167
  • Pillai RS, Bhattacharyya SN, Filipowicz W. Repression of protein synthesis by miRnas: how many mechanisms? Trends Cell Biol. 2007;17(3):118–126. doi: 10.1016/j.tcb.2006.12.007
  • Hong TH, Park IY. MicroRNA expression profiling of diagnostic needle aspirates from surgical pancreatic cancer specimens. Ann Surg Treat Res. 2014;87:290–297. doi: 10.4174/astr.2014.87.6.290
  • Hampton T. MicroRNAs linked to pancreatic cancer. JAMA. 2007;297:937.
  • Ali A, Jamieson NB, Khan IN, et al. Prognostic implications of microRNA-21 overexpression in pancreatic ductal adenocarcinoma: an international multicenter study of 686 patients. Am J Cancer Res. 2022;12:5668–5683.
  • Frampton AE, Castellano L, Colombo T, et al. Integrated molecular analysis to investigate the role of microRnas in pancreatic tumour growth and progression. Lancet Lond Engl. 2015;385(Suppl 1):S37. doi: 10.1016/S0140-6736(15)60352-X
  • Giovannetti E, Funel N, Peters GJ, et al. MicroRNA-21 in pancreatic cancer: correlation with clinical outcome and pharmacologic aspects underlying its role in the modulation of gemcitabine activity. Cancer Res. 2010;70:4528–4538. doi: 10.1158/0008-5472.CAN-09-4467
  • Vahabi M, Dehni B, Antomás I, et al. Targeting miRNA and using miRNA as potential therapeutic options to bypass resistance in pancreatic ductal adenocarcinoma. Cancer Metastasis Rev. 2023;42:725–740. doi: 10.1007/s10555-023-10127-w
  • Mittal V. Epithelial mesenchymal transition in tumor metastasis. Annu Rev Pathol. 2018;13:395–412. doi: 10.1146/annurev-pathol-020117-043854
  • El Amrani M, Corfiotti F, Corvaisier M, et al. Gemcitabine-induced epithelial-mesenchymal transition-like changes sustain chemoresistance of pancreatic cancer cells of mesenchymal-like phenotype. Mol Carcinog. 2019;58:1985–1997. doi: 10.1002/mc.23090
  • Kuwada K, Kagawa S, Yoshida R, et al. The epithelial-to-mesenchymal transition induced by tumor-associated macrophages confers chemoresistance in peritoneally disseminated pancreatic cancer. J Exp Clin Cancer Res. 2018;37:307. doi: 10.1186/s13046-018-0981-2
  • Hwang JS, Lai TH, Ahmed M, et al. Regulation of TGF-β1-induced EMT by autophagy-dependent energy metabolism in cancer cells. Cancers (Basel). 2022;14:4845. doi: 10.3390/cancers14194845
  • Izumchenko E, Chang X, Michailidi C, et al. The TGFβ–miR200–MIG6 pathway orchestrates the EMT-Associated kinase switch that induces resistance to EGFR inhibitors. Cancer Res. 2014;74:3995–4005. doi: 10.1158/0008-5472.CAN-14-0110
  • Sanomachi T, Suzuki S, Togashi K, et al. Brexpiprazole reduces survivin and reverses EGFR tyrosine kinase inhibitor resistance in lung and pancreatic cancer. Anticancer Res. 2019;39:4817–4828. doi: 10.21873/anticanres.13667
  • Zhao X, Yang Y, Yu H, et al. Polydatin inhibits ZEB1-invoked epithelial-mesenchymal transition in fructose-induced liver fibrosis. J Cell Mol Med. 2020;24:13208–13222. doi: 10.1111/jcmm.15933
  • Melchionna R, Iapicca P, Di Modugno F, et al. The pattern of hMENA isoforms is regulated by TGF-β1 in pancreatic cancer and may predict patient outcome. Oncoimmunology. 2016;5:e1221556. doi: 10.1080/2162402X.2016.1221556
  • Randazzo O, Papini F, Mantini G, et al. “Open Sesame?”: biomarker status of the human equilibrative nucleoside transporter-1 and molecular mechanisms influencing its expression and activity in the uptake and cytotoxicity of gemcitabine in pancreatic cancer. Cancers (Basel). 2020;12:3206. doi: 10.3390/cancers12113206
  • Toledo B, González-Titos A, Hernández-Camarero P, et al. A Brief Review on chemoresistance; targeting cancer stem cells as an alternative approach. Int J Mol Sci. 2023;24:4487. doi: 10.3390/ijms24054487
  • Wang C, Lin W, Playa H, et al. Dipyridamole analogs as pharmacological inhibitors of equilibrative nucleoside transporters. Identification of novel potent and selective inhibitors of the adenosine transporter function of human equilibrative nucleoside transporter 4 (hENT4). Biochem Pharmacol. 2013;86:1531–1540. doi: 10.1016/j.bcp.2013.08.063
  • Farrell JJ, Elsaleh H, Garcia M, et al. Human equilibrative nucleoside transporter 1 levels predict response to gemcitabine in patients with pancreatic cancer. Gastroenterology. 2009;136:187–195. doi: 10.1053/j.gastro.2008.09.067
  • Greenhalf W, Ghaneh P, Neoptolemos JP, et al. Pancreatic cancer hENT1 expression and survival from gemcitabine in patients from the ESPAC-3 trial. JNCI J Natl Cancer Inst. 2014;106:djt347. doi: 10.1093/jnci/djt347
  • Hioki M, Shimada T, Yuan T, et al. Contribution of equilibrative nucleoside transporters 1 and 2 to gemcitabine uptake in pancreatic cancer cells. Biopharm Drug Dispos. 2018;39:256–264. doi: 10.1002/bdd.2131
  • Paproski RJ, Young JD, Cass CE. Predicting gemcitabine transport and toxicity in human pancreatic cancer cell lines with the positron emission tomography tracer 3′-deoxy-3′-fluorothymidine. Biochem Pharmacol. 2010;79:587–595. doi: 10.1016/j.bcp.2009.09.025
  • Lockhart AC, Tirona RG, Kim RB. Pharmacogenetics of ATP-binding cassette transporters in cancer and chemotherapy. Mol Cancer Ther. 2003;2:685–698.
  • Mathijssen RHJ, Marsh S, Karlsson MO, et al. Irinotecan pathway genotype analysis to predict pharmacokinetics. Clin Cancer Res Off J Am Assoc Cancer Res. 2003;9:3246–3253.
  • Chen M, Xue X, Wang F, et al. Expression and promoter methylation analysis of ATP-binding cassette genes in pancreatic cancer. Oncol Rep. 2012;27:265–269. doi: 10.3892/or.2011.1475
  • Yao L, Gu J, Mao Y, et al. Dynamic quantitative detection of ABC transporter family promoter methylation by MS‑HRM for predicting MDR in pancreatic cancer. Oncol Lett. 2018;15:5602–5610. doi: 10.3892/ol.2018.8041
  • Yada E, Kasajima R, Niida A, et al. Possible role of cytochrome P450 1B1 in the mechanism of gemcitabine resistance in pancreatic cancer. Biomedicines. 2021;9:1396. doi: 10.3390/biomedicines9101396
  • Leskelä S, Jara C, Leandro-García LJ, et al. Polymorphisms in cytochromes P450 2C8 and 3A5 are associated with paclitaxel neurotoxicity. Pharmacogenomics J. 2011;11:121–129. doi: 10.1038/tpj.2010.13
  • Bergonzini C, Gregori A, Hagens TMS, et al. ABCB1 overexpression through locus amplification represents an actionable target to combat paclitaxel resistance in pancreatic cancer cells [internet].bioRxiv; 2023 [cited 2024 Jan 26]. p. 2023.05.30.542412. Available from: https://www.biorxiv.org/content/10.1101/2023.05.30.542412v1
  • Neuzillet C, Tijeras-Raballand A, Cros J, et al. Stromal expression of SPARC in pancreatic adenocarcinoma. Cancer Metastasis Rev. 2013;32:585–602. doi: 10.1007/s10555-013-9439-3
  • Von Hoff DD, Ramanathan RK, Borad MJ, et al. Gemcitabine plus nab-Paclitaxel is an active regimen in patients with advanced pancreatic cancer: a phase I/II trial. J Clin Oncol. 2011;29(34):4548–4554. doi: 10.1200/JCO.2011.36.5742
  • Tuveson DA Data from nab-paclitaxel potentiates gemcitabine activity by reducing cytidine deaminase levels in a mouse Model of pancreatic cancer. 2023 [cited 2023 Nov 15]. Available from: https://aacr.figshare.com/collections/Data_from_i_nab_i_-Paclitaxel_Potentiates_Gemcitabine_Activity_by_Reducing_Cytidine_Deaminase_Levels_in_a_Mouse_Model_of_Pancreatic_Cancer/6546821
  • Thomas D, Radhakrishnan P. Tumor-stromal crosstalk in pancreatic cancer and tissue fibrosis. Mol Cancer. 2019;18:14. doi: 10.1186/s12943-018-0927-5
  • Liot S, Balas J, Aubert A, et al. Stroma involvement in pancreatic ductal adenocarcinoma: an overview focusing on extracellular matrix proteins. Front Immunol. 2021;12 [cited 2023 Nov 15]. [Internet] Available from: https://www.frontiersin.org/articles/10.3389/fimmu.2021.612271
  • Liang C, Shi S, Meng Q, et al. Complex roles of the stroma in the intrinsic resistance to gemcitabine in pancreatic cancer: where we are and where we are going. Exp Mol Med. 2017;49:e406. doi: 10.1038/emm.2017.255
  • Rhim AD, Oberstein PE, Thomas DH, et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 2014;25:735–747. doi: 10.1016/j.ccr.2014.04.021
  • Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013
  • Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013;19:1423–1437. doi: 10.1038/nm.3394
  • Neesse A, Algül H, Tuveson DA, et al. Stromal biology and therapy in pancreatic cancer: a changing paradigm. Gut. 2015;64:1476–1484. doi: 10.1136/gutjnl-2015-309304
  • Grasso C, Jansen G, Giovannetti E. Drug resistance in pancreatic cancer: impact of altered energy metabolism. Crit Rev Oncol Hematol. 2017;114:139–152. doi: 10.1016/j.critrevonc.2017.03.026
  • Poklepovic AS, Fields EC, Bandyopadhyay D, et al. A phase 1 study of neoadjuvant chemotherapy followed by concurrent chemoradiation with gemcitabine, sorafenib, and vorinostat in pancreatic cancer. J Clin Oncol. 2021;39:e16268–e16268. doi: 10.1200/JCO.2021.39.15_suppl.e16268
  • Algaze S, Hanna DL, Azad NS, et al. A phase Ib study of guadecitabine and durvalumab in patients with advanced hepatocellular carcinoma, pancreatic adenocarcinoma, and biliary cancers. J Clin Oncol. 2022;40:574–574. doi: 10.1200/JCO.2022.40.4_suppl.574
  • Barbato A, Piscopo F, Salati M, et al. Micro-RNA in cholangiocarcinoma: implications for diagnosis, prognosis, and therapy. J Mol Pathol. 2022;3:88–103. doi: 10.3390/jmp3020009
  • Xie Y, Wang Y, Li J, et al. Cholangiocarcinoma therapy with nanoparticles that combine downregulation of MicroRNA-210 with inhibition of cancer cell invasiveness. Theranostics. 2018;8:4305. doi: 10.7150/thno.26506
  • Beg MS, Brenner AJ, Sachdev J, et al. Phase I study of MRX34, a liposomal miR-34a mimic, administered twice weekly in patients with advanced solid tumors. Invest New Drugs. 2017;35:180–188. doi: 10.1007/s10637-016-0407-y
  • Yen W-C, Fischer MM, Axelrod F, et al. Targeting notch signaling with a Notch2/Notch3 antagonist (tarextumab) inhibits tumor growth and decreases tumor-initiating cell frequency. Clin Cancer Res. 2015;21:2084–2095. doi: 10.1158/1078-0432.CCR-14-2808
  • O’Reilly EM, Sahai V, Bendell JC, et al. Results of a randomized phase II trial of an anti-notch 2/3, tarextumab (OMP-59R5, TRXT, anti-Notch2/3), in combination with nab-paclitaxel and gemcitabine (nab-P+Gem) in patients (pts) with untreated metastatic pancreatic cancer (mPC). J Clin Oncol. 2017;35:279–279. doi: 10.1200/JCO.2017.35.4_suppl.279
  • Cook N, Basu B, Smith D-M, et al. A phase I trial of the ɣ-secretase inhibitor (GSI) MK-0752 in combination with gemcitabine in patients with pancreatic ductal adenocarcinoma (PDAC). J Clin Oncol. 2014;32:4116–4116. doi: 10.1200/jco.2014.32.15_suppl.4116
  • Gulla A, Andriusaityte U, Zdanys GT, et al. The impact of epithelial–mesenchymal transition and metformin on pancreatic cancer chemoresistance: a pathway towards individualized therapy. Medicina (Mex). 2022;58:467. doi: 10.3390/medicina58040467
  • Glabman RA, Choyke PL, Sato N. Cancer-associated fibroblasts: tumorigenicity and targeting for cancer therapy. Cancers (Basel). 2022;14:3906. doi: 10.3390/cancers14163906
  • Albrengues J, Bertero T, Grasset E, et al. Epigenetic switch drives the conversion of fibroblasts into proinvasive cancer-associated fibroblasts. Nat Commun. 2015;6:10204. doi: 10.1038/ncomms10204
  • Gailhouste L, Liew LC, Hatada I, et al. Epigenetic reprogramming using 5-azacytidine promotes an anti-cancer response in pancreatic adenocarcinoma cells. Cell Death Dis. 2018;9:1–12. doi: 10.1038/s41419-018-0487-z
  • Safyan RA, Manji GA, Lee SM, et al. Phase 2 study of azacitidine (AZA) plus pembrolizumab (pembro) as second-line treatment in patients with advanced pancreatic ductal adenocarcinoma. J Clin Oncol. 2022;40:4158–4158. doi: 10.1200/JCO.2022.40.16_suppl.4158
  • Jimeno A, Weiss GJ, Miller WH, et al. Phase I study of the hedgehog pathway inhibitor IPI-926 in adult patients with solid tumors. Clin Cancer Res Off J Am Assoc Cancer Res. 2013;19:2766–2774. doi: 10.1158/1078-0432.CCR-12-3654
  • Hingorani SR, Zheng L, Bullock AJ, et al. HALO 202: randomized phase II study of PEGPH20 plus nab-Paclitaxel/Gemcitabine versus nab-Paclitaxel/Gemcitabine in patients with untreated, metastatic pancreatic ductal adenocarcinoma. J Clin Oncol. 2018;36:359–366. doi: 10.1200/JCO.2017.74.9564
  • Ramanathan RK, McDonough SL, Philip PA, et al. Phase IB/II randomized study of FOLFIRINOX plus pegylated recombinant human hyaluronidase versus FOLFIRINOX alone in patients with metastatic pancreatic adenocarcinoma: SWOG S1313. J Clin Oncol. 2019;37:1062–1069. doi: 10.1200/JCO.18.01295
  • Hurwitz HI, Uppal N, Wagner SA, et al. Randomized, double-blind, phase II study of ruxolitinib or placebo in combination with capecitabine in patients with metastatic pancreatic cancer for whom therapy with gemcitabine has failed. J Clin Oncol. 2015;33:4039–4047. doi: 10.1200/JCO.2015.61.4578
  • Hurwitz H, Van Cutsem E, Bendell JC, et al. Two randomized, placebo-controlled phase 3 studies of ruxolitinib (rux) + capecitabine (C) in patients (pts) with advanced/metastatic pancreatic cancer (mPC) after failure/intolerance of first-line chemotherapy: JANUS 1 (J1) and JANUS 2 (J2). J Clin Oncol. 2017;35:343–343. doi: 10.1200/JCO.2017.35.4_suppl.343
  • Valle J, Wasan H, Palmer DH, et al. Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. N Engl J Med. 2010;362:1273–1281. doi: 10.1056/NEJMoa0908721
  • Shroff RT, Javle MM, Xiao L, et al. Gemcitabine, cisplatin, and nab-paclitaxel for the treatment of advanced biliary tract cancers: a phase 2 clinical trial. JAMA Oncol. 2019;5:824–830. doi: 10.1001/jamaoncol.2019.0270
  • SWOG Cancer Research Network. A phase III randomized trial of Gemcitabine, cisplatin, and nab-paclitaxel versus Gemcitabine and cisplatin in newly diagnosed, advanced biliary tract cancers [Internet]. Report No.: NCT03768414. clinicaltrials.gov; 2023 [cited 2024 Jan 1]. Available from: https://clinicaltrials.gov/study/NCT03768414
  • Lopes LF, Bacchi CE. Imatinib treatment for gastrointestinal stromal tumour (GIST). J Cell Mol Med. 2010;14:42–50. doi: 10.1111/j.1582-4934.2009.00983.x
  • Soria J-C, Ohe Y, Vansteenkiste J, et al. Osimertinib in untreated EGFR-mutated advanced non–small-cell lung cancer. N Engl J Med. 2018;378:113–125. doi: 10.1056/NEJMoa1713137
  • Sabnis AJ, Bivona TG. Principles of resistance to targeted cancer therapy: lessons from basic and translational cancer biology. Trends Mol Med. 2019;25:185–197. doi: 10.1016/j.molmed.2018.12.009
  • Yun C-H, Mengwasser KE, Toms AV, et al. The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP. Proc Natl Acad Sci. 2008;105:2070–2075. doi: 10.1073/pnas.0709662105
  • Poulikakos PI, Persaud Y, Janakiraman M, et al. RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF(V600E). Nature. 2011;480:387–390. doi: 10.1038/nature10662
  • Gorre ME, Mohammed M, Ellwood K, et al. Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification. Science. 2001;293:876–880. doi: 10.1126/science.1062538
  • Nazarian R, Shi H, Wang Q, et al. Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature. 2010;468:973–977. doi: 10.1038/nature09626
  • Coleman N, Hong L, Zhang J, et al. Beyond epidermal growth factor receptor: MET amplification as a general resistance driver to targeted therapy in oncogene-driven non-small-cell lung cancer. ESMO Open. 2021;6:100319. doi: 10.1016/j.esmoop.2021.100319
  • De Las Rivas J, Brozovic A, Izraely S, et al. Cancer drug resistance induced by EMT: novel therapeutic strategies. Arch Toxicol. 2021;95:2279–2297. doi: 10.1007/s00204-021-03063-7
  • Yin X, Li Y, Wang H, et al. Small cell lung cancer transformation: From pathogenesis to treatment. Semin Cancer Biol. 2022;86:595–606. doi: 10.1016/j.semcancer.2022.03.006
  • Yang J, Antin P, Berx G, et al. Guidelines and definitions for research on epithelial–mesenchymal transition. Nat Rev Mol Cell Biol. 2020;21:341–352. doi: 10.1038/s41580-020-0237-9
  • Varghese AM, Patel JAA, Janjigian YY, et al. Non-invasive detection of acquired resistance to FGFR inhibition in patients with cholangiocarcinoma harboring FGFR2 alterations. JCO. 2019;37(15_suppl):4096–4096. doi: 10.1200/JCO.2019.37.15_suppl.4096
  • Goyal L, Saha SK, Liu LY, et al. Polyclonal secondary FGFR2 mutations drive acquired resistance to FGFR inhibition in patients with FGFR2 fusion–positive cholangiocarcinoma. Cancer Discov. 2017;7:252–263. doi: 10.1158/2159-8290.CD-16-1000
  • Silverman IM, Hollebecque A, Friboulet L, et al. Clinicogenomic analysis of FGFR2-rearranged cholangiocarcinoma identifies correlates of response and mechanisms of resistance to pemigatinib. Cancer Discov. 2021;11:326–339. doi: 10.1158/2159-8290.CD-20-0766
  • Lubner SJ, Mahoney MR, Kolesar JL, et al. Report of a multicenter phase II trial testing a combination of biweekly bevacizumab and daily erlotinib in patients with unresectable biliary cancer: a phase II consortium study. J Clin Oncol. 2010;28:3491–3497. doi: 10.1200/JCO.2010.28.4075
  • Wu Q, Zhen Y, Shi L, et al. EGFR inhibition potentiates FGFR inhibitor therapy and overcomes resistance in FGFR2 fusion–positive cholangiocarcinoma. Cancer Discov. 2022;12:1378–1395. doi: 10.1158/2159-8290.CD-21-1168
  • Krook MA, Lenyo A, Wilberding M, et al. Efficacy of FGFR inhibitors and combination therapies for acquired resistance in FGFR2-fusion cholangiocarcinoma. Mol Cancer Ther. 2020;19:847–857. doi: 10.1158/1535-7163.MCT-19-0631
  • Vaquero J, Guedj N, Clapéron A, et al. Epithelial-mesenchymal transition in cholangiocarcinoma: from clinical evidence to regulatory networks. J Hepatol. 2017;66:424–441. doi: 10.1016/j.jhep.2016.09.010
  • Vaquero J, Lobe C, Tahraoui S, et al. The IGF2/IR/IGF1R Pathway in tumor Cells and myofibroblasts mediates resistance to EGFR inhibition in cholangiocarcinoma. Clin Cancer Res Off J Am Assoc Cancer Res. 2018;24:4282–4296. doi: 10.1158/1078-0432.CCR-17-3725
  • Hu ZI, O’Reilly EM. Therapeutic developments in pancreatic cancer. Nat Rev Gastroenterol Hepatol. 2023;21(1):7–24. doi: 10.1038/s41575-023-00840-w
  • Gurreri E, Genovese G, Perelli L, et al. KRAS-Dependency in pancreatic ductal adenocarcinoma: mechanisms of escaping in resistance to KRAS inhibitors and perspectives of therapy. Int J Mol Sci. 2023;24:9313.
  • Qin S, Bai Y, Wang Z, et al. Nimotuzumab combined with gemcitabine versus gemcitabine in K-RAS wild-type locally advanced or metastatic pancreatic cancer: a prospective, randomized-controlled, double-blinded, multicenter, and phase III clinical trial. J Clin Oncol. 2022;40:LBA4011–LBA4011. doi: 10.1200/JCO.2022.40.17_suppl.LBA4011
  • Qian Y, Gong Y, Fan Z, et al. Molecular alterations and targeted therapy in pancreatic ductal adenocarcinoma. J Hematol OncolJ Hematol Oncol. 2020;13:130. doi: 10.1186/s13045-020-00958-3
  • Gout J, Perkhofer L, Morawe M, et al. Synergistic targeting and resistance to PARP inhibition in DNA damage repair-deficient pancreatic cancer. Gut. 2021;70:743–760. doi: 10.1136/gutjnl-2019-319970
  • Ettrich TJ, Schwerdel D, Dolnik A, et al. Genotyping of circulating tumor DNA in cholangiocarcinoma reveals diagnostic and prognostic information. Sci Rep. 2019;9:13261. doi: 10.1038/s41598-019-49860-0
  • Luchini C, Veronese N, Nottegar A, et al. Liquid biopsy as surrogate for tissue for molecular profiling in pancreatic cancer: a meta-analysis towards precision medicine. Cancers (Basel). 2019;11:1152. doi: 10.3390/cancers11081152
  • Gray JE, Okamoto I, Sriuranpong V, et al. Tissue and plasma EGFR mutation analysis in the FLAURA Trial: osimertinib versus Comparator EGFR Tyrosine Kinase Inhibitor as first-line treatment in patients with EGFR-Mutated advanced non–small cell lung cancer. Clin Cancer Res. 2019;25:6644–6652. doi: 10.1158/1078-0432.CCR-19-1126
  • Goyal L, Shi L, Liu LY, et al. TAS-120 overcomes resistance to ATP-competitive FGFR inhibitors in patients with FGFR2 fusion–positive Intrahepatic cholangiocarcinoma. Cancer Discov. 2019;9:1064–1079. doi: 10.1158/2159-8290.CD-19-0182
  • Pant S, Schuler MH, Iyer G, et al. Efficacy and safety of erdafitinib in adults with cholangiocarcinoma (CCA) with prespecified fibroblast growth factor receptor alterations (FGFRalt) in the phase 2 open-label, single-arm RAGNAR trial: expansion cohort results. J Clin Oncol. 2023;41:610–610. doi: 10.1200/JCO.2023.41.4_suppl.610
  • Javle MM, Fountzilas C, Li D, et al. Phase II study of FGFR1-3 inhibitor tinengotinib as monotherapy in patients with advanced or metastatic cholangiocarcinoma: Interim analysis. J Clin Oncol. 2023;41:539–539. doi: 10.1200/JCO.2023.41.4_suppl.539
  • Wang X, Allen S, Blake JF, et al. Identification of MRTX1133, a noncovalent, potent, and selective KRASG12D inhibitor. J Med Chem. 2022;65:3123–3133. doi: 10.1021/acs.jmedchem.1c01688
  • Mirati Therapeutics Inc. A phase 1/2 multiple expansion cohort trial of MRTX1133 in patients with advanced solid tumors harboring a KRAS G12D mutation [internet]. Report No.: NCT05737706. clinicaltrials.gov; 2023 [cited 2023 Jan 1]. Available from: https://clinicaltrials.gov/study/NCT05737706
  • Hofmann MH, Gmachl M, Ramharter J, et al. BI-3406, a potent and selective SOS1–KRAS interaction inhibitor, is effective in KRAS-Driven cancers through combined MEK inhibition. Cancer Discov. 2021;11:142–157. doi: 10.1158/2159-8290.CD-20-0142
  • Revolution medicines, Inc. A multicenter open-label study of RMC-6236 in patients with advanced solid tumors harboring specific mutations in RAS [internet]. Report No.: NCT05379985. clinicaltrials.gov; 2023 [cited 2023 Jan 1]. Available from: https://clinicaltrials.gov/study/NCT05379985
  • Revolution Medicines, Inc. Phase 1/1b, multicenter, open-label, dose escalation and dose expansion study of RMC-6291 monotherapy in subjects with advanced KRASG12C mutant solid tumors [Internet]. Report No.: NCT05462717. clinicaltrials.gov; 2023 [cited 2023Jan 1]. Available from: https://clinicaltrials.gov/study/NCT05462717
  • Brana I, Shapiro G, Johnson ML, et al. Initial results from a dose finding study of TNO155, a SHP2 inhibitor, in adults with advanced solid tumors. J Clin Oncol. 2021;39:3005–3005. doi: 10.1200/JCO.2021.39.15_suppl.3005
  • Krishnan T, Roberts-Thomson R, Broadbridge V, et al. Targeting mutated KRAS genes to treat solid tumours. Mol Diagn Ther. 2022;26:39–49. doi: 10.1007/s40291-021-00564-0
  • Wallez Y, Dunlop CR, Johnson TI, et al. The ATR inhibitor AZD6738 synergizes with gemcitabine in vitro and in vivo to induce pancreatic ductal adenocarcinoma regression. Mol Cancer Ther. 2018;17:1670–1682. doi: 10.1158/1535-7163.MCT-18-0010
  • Prevo R, Fokas E, Reaper PM, et al. The novel ATR inhibitor VE-821 increases sensitivity of pancreatic cancer cells to radiation and chemotherapy. Cancer Biol Ther. 2012;13:1072–1081. doi: 10.4161/cbt.21093
  • Long GV, Stroyakovskiy D, Gogas H, et al. Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: a multicentre, double-blind, phase 3 randomised controlled trial. Lancet. 2015;386:444–451. doi: 10.1016/S0140-6736(15)60898-4
  • Brown WS, McDonald PC, Nemirovsky O, et al. Overcoming adaptive resistance to KRAS and MEK inhibitors by Co-targeting mTORC1/2 complexes in pancreatic cancer. Cell Rep Med [Internet]. 2020;1 [cited 2023 Oct 16]. Available from: https://www.cell.com/cell-reports-medicine/abstract/S2666-3791(20)30173-7
  • Bannoura SF, Uddin M, Nagasaka M, et al. Targeting KRAS in pancreatic cancer: new drugs on the horizon. Cancer Metastasis Rev. 2021;40:819–835. doi: 10.1007/s10555-021-09990-2
  • LoRusso P, Fakih M, FYFLD V, et al. 561P phase ib study of ribociclib (R) + trametinib (T) in patients (pts) with metastatic/advanced solid tumours. Ann Oncol. 2020;31:S484. doi: 10.1016/j.annonc.2020.08.675
  • Wang J, Zhao J, Zhong J, et al. 653O glecirasib (KRAS G12C inhibitor) in combination with JAB-3312 (SHP2 inhibitor) in patients with KRAS p.G12C mutated solid tumors. Ann Oncol. 2023;34:S459. doi: 10.1016/j.annonc.2023.09.1839
  • BU C, ZHAO L, WANG L, et al. mTORC2 promotes pancreatic cancer progression and parp inhibitor resistance. Oncol Res. 31:495–503. doi: 10.32604/or.2023.029309
  • Rose M, Burgess JT, O’Byrne K, et al. PARP inhibitors: clinical relevance, mechanisms of action and tumor resistance. Front Cell Dev Biol. 2020;8:564601. doi: 10.3389/fcell.2020.564601
  • Revythis A, Limbu A, Mikropoulos C, et al. Recent insights into PARP and immuno-checkpoint inhibitors in Epithelial ovarian cancer. Int J Environ Res Public Health. 2022;19:8577. doi: 10.3390/ijerph19148577
  • Reiss KA, Mick R, Teitelbaum U, et al. Niraparib plus nivolumab or niraparib plus ipilimumab in patients with platinum-sensitive advanced pancreatic cancer: a randomised, phase 1b/2 trial. Lancet Oncol. 2022;23:1009–1020. doi: 10.1016/S1470-2045(22)00369-2
  • Corrie PG. A phase II study combining pembrolizumab with olaparib in metastatic pancreatic adenocarcinoma (PDA) patients with high Tumour Mutation Burden [internet]. Report No.: NCT05093231. clinicaltrials.gov; 2021 [cited 2024 Jan 1]. Available from: https://clinicaltrials.gov/study/NCT05093231
  • National Cancer Institute (NCI). Randomized phase II clinical trial of olaparib + pembrolizumab vs. Olaparib alone as maintenance therapy in metastatic pancreatic cancer patients with germline BRCA1 or BRCA2 mutations [internet]. Report No.: NCT04548752. clinicaltrials.gov; 2024 [cited 2024 Jan 1]. Available from: https://clinicaltrials.gov/study/NCT04548752
  • Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins. Multi-agent Low Dose Chemotherapy (gemcitabine, Nab-paclitaxel, capecitabine, cisplatin, Irinotecan) followed by maintenance olaparib and Pembrolizumab in untreated metastatic pancreatic ductal adenocarcinoma [internet]. Report No.: NCT04753879. clinicaltrials.gov; 2023 [cited 2023 Jan 1]. Available from: https://clinicaltrials.gov/study/NCT04753879
  • Mayo Clinic. Phase II study of Niraparib and TSR-042 in Patients with Germline or Somatic BRCA1/2 and PALB2-Related pancreatic cancer [internet]. Report No.: NCT04493060. clinicaltrials.gov; 2023 [cited 2023 Jan 1]. Available from: https://clinicaltrials.gov/study/NCT04493060
  • Servier Bio-Innovation LLC. A phase 1/2, Safety Lead-in and dose expansion, open-label, multicenter trial investigating the Safety, tolerability, and preliminary activity of Ivosidenib in combination with nivolumab and Ipilimumab in Previously treated subjects with nonresectable or metastatic cholangiocarcinoma with an IDH1 Mutation [internet]. Report No.: NCT05921760.clinicaltrials.gov; 2023 [cited 2024 Jan 1]. Available from: https://clinicaltrials.gov/study/NCT05921760
  • University Health Network, Toronto. A phase II study of Olaparib and durvalumab (MEDI 4736) in patients with IDH-Mutated solid tumors [internet]. Report No.: NCT03991832. clinicaltrials.gov; 2023 [cited 2024 Jan 1]. Available from: https://clinicaltrials.gov/study/NCT03991832
  • Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359:1350–1355. doi: 10.1126/science.aar4060
  • Said SS, Ibrahim WN. Cancer resistance to immunotherapy: comprehensive insights with future perspectives. Pharmaceutics. 2023;15:1143. doi: 10.3390/pharmaceutics15041143
  • Schmiechen ZC, Stromnes IM. Mechanisms governing immunotherapy resistance in pancreatic ductal adenocarcinoma. Front Immunol [Internet]. 2021 [cited 2023 Oct 20];11. doi: 10.3389/fimmu.2020.613815
  • Yoon JH, Jung Y-J, Moon S-H. Immunotherapy for pancreatic cancer. World J Clin Cases. 2021;9:2969–2982. doi: 10.12998/wjcc.v9.i13.2969
  • Nakamura H, Arai Y, Totoki Y, et al. Genomic spectra of biliary tract cancer. Nat Genet. 2015;47:1003–1010. doi: 10.1038/ng.3375
  • Job S, Rapoud D, Dos Santos A, et al. Identification of four immune subtypes characterized by distinct composition and functions of tumor microenvironment in intrahepatic cholangiocarcinoma. Hepatology. 2020;72:965. doi: 10.1002/hep.31092
  • Lo JH, Agarwal R, Goff LW, et al. Immunotherapy in biliary tract cancers: current standard-of-care and emerging strategies. Cancers (Basel). 2023;15:3312. doi: 10.3390/cancers15133312
  • Goeppert B, Frauenschuh L, Zucknick M, et al. Major histocompatibility complex class I expression impacts on patient survival and type and density of immune cells in biliary tract cancer. Br J Cancer. 2015;113:1343–1349. doi: 10.1038/bjc.2015.337
  • Amankulor NM, Kim Y, Arora S, et al. Mutant IDH1 regulates the tumor-associated immune system in gliomas. Genes Dev. 2017;31:774–786. doi: 10.1101/gad.294991.116
  • Ruan R, Li L, Li X, et al. Unleashing the potential of combining FGFR inhibitor and immune checkpoint blockade for FGF/FGFR signaling in tumor microenvironment. Mol Cancer. 2023;22:60. doi: 10.1186/s12943-023-01761-7
  • Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409–413. doi: 10.1126/science.aan6733
  • Heumann T, Azad N. Next-generation immunotherapy for pancreatic ductal adenocarcinoma: navigating pathways of immune resistance. Cancer Metastasis Rev. 2021;40:837–862. doi: 10.1007/s10555-021-09981-3
  • Feig C, Gopinathan A, Neesse A, et al. The pancreas cancer microenvironment. Clin Cancer Res Off J Am Assoc Cancer Res. 2012;18:4266–4276. doi: 10.1158/1078-0432.CCR-11-3114
  • Knudsen ES, Vail P, Balaji U, et al. Stratification of pancreatic ductal adenocarcinoma: combinatorial genetic, stromal, and immunological markers. Clin Cancer Res Off J Am Assoc Cancer Res. 2017;23:4429. doi: 10.1158/1078-0432.CCR-17-0162
  • Piha-Paul SA, Oh D-Y, Ueno M, et al. Efficacy and safety of pembrolizumab for the treatment of advanced biliary cancer: results from the KEYNOTE-158 and KEYNOTE-028 studies. Int J Cancer. 2020;147:2190–2198. doi: 10.1002/ijc.33013
  • Kim RD, Chung V, Alese OB, et al. A phase 2 multi-institutional study of nivolumab for patients with advanced refractory biliary tract cancer. JAMA Oncol. 2020;6:888–894. doi: 10.1001/jamaoncol.2020.0930
  • Terrero G, Datta J, Dennison J, et al. Ipilimumab/Nivolumab therapy in patients with metastatic pancreatic or biliary cancer with homologous recombination deficiency pathogenic germline variants. JAMA Oncol. 2022;8:938–940. doi: 10.1001/jamaoncol.2022.0611
  • Klein O, Kee D, Nagrial A, et al. Evaluation of combination nivolumab and ipilimumab immunotherapy in patients with advanced biliary tract cancers: subgroup analysis of a phase 2 nonrandomized clinical trial. JAMA Oncol. 2020;6:1405–1409. doi: 10.1001/jamaoncol.2020.2814
  • Doki Y, Ueno M, Hsu C-H, 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. 2022;11:2550–2560. doi: 10.1002/cam4.4593
  • Lin J, Yang X, Long J, et al. Pembrolizumab combined with lenvatinib as non-first-line therapy in patients with refractory biliary tract carcinoma. Hepatobiliary Surg Nutr. 2020;9:41424–41424. doi: 10.21037/hbsn-20-338
  • Cousin S, Cantarel C, Guegan J-P, et al. Regorafenib–avelumab combination in patients with biliary tract cancer (REGOMUNE): a single-arm, open-label, phase II trial. Eur J Cancer. 2022;162:161–169. doi: 10.1016/j.ejca.2021.11.012
  • Shi G-M, Huang X-Y, Wu D, 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;8:1–10. doi: 10.1038/s41392-023-01317-7
  • Guo Y, Feng K, Liu Y, et al. Phase I study of Chimeric Antigen Receptor–modified T cells in patients with EGFR-Positive advanced biliary tract cancers. Clin Cancer Res. 2018;24:1277–1286. doi: 10.1158/1078-0432.CCR-17-0432
  • Koido S, Homma S, Okamoto M, et al. Chemoimmunotherapy targeting Wilms’ tumor 1 (WT1)-specific cytotoxic T lymphocyte and helper T cell responses for patients with pancreatic cancer. Oncoimmunology. 2014;3:e958950. doi: 10.4161/21624011.2014.958950
  • Wang-Gillam A, Plambeck-Suess S, Goedegebuure P, et al. A phase I study of IMP321 and gemcitabine as the front-line therapy in patients with advanced pancreatic adenocarcinoma. Invest New Drugs. 2013;31:707–713. doi: 10.1007/s10637-012-9866-y
  • Bahary N, Wang-Gillam A, Haraldsdottir S, et al. Phase 2 trial of the IDO pathway inhibitor indoximod plus gemcitabine/nab-paclitaxel for the treatment of patients with metastatic pancreas cancer. J Clin Oncol. 2018;36:4015–4015. doi: 10.1200/JCO.2018.36.15_suppl.4015
  • Marin-Acevedo JA, Dholaria B, Soyano AE, et al. Next generation of immune checkpoint therapy in cancer: new developments and challenges. J Hematol OncolJ Hematol Oncol. 2018;11:39. doi: 10.1186/s13045-018-0582-8
  • Rojas LA, Sethna Z, Soares KC, et al. Personalized RNA neoantigen vaccines stimulate T cells in pancreatic cancer. Nature. 2023;618:144–150. doi: 10.1038/s41586-023-06063-y
  • Feng Q, Sun B, Xue T, et al. Advances in CAR T-cell therapy in bile duct, pancreatic, and gastric cancers. Front Immunol. 2022;13 [cited 2023 Oct 20]. [Internet] Available from: https://www.frontiersin.org/articles/10.3389/fimmu.2022.1025608
  • Leem G, Jang S-I, Cho J-H, et al. Safety and efficacy of allogeneic natural killer cells in combination with pembrolizumab in patients with chemotherapy-refractory biliary tract cancer: a multicenter open-label phase 1/2a trial. Cancers (Basel). 2022;14:4229. doi: 10.3390/cancers14174229
  • Elrakaybi A, Ruess DA, Lübbert M, et al. Epigenetics in Pancreatic Ductal Adenocarcinoma: Impact on Biology and Utilization in Diagnostics and Treatment. Cancers (Basel). 2022;14:5926. doi: 10.3390/cancers14235926
  • Wang L Capecitabine combined with lenvatinib and Tislelizumab as adjuvant treatment after resection in patients with biliary tract cancer: a single-arm, phase II study [internet]. Report No.: NCT05254847. clinicaltrials.gov; 2022 [cited 2024 Jan 1]. Available from: https://clinicaltrials.gov/study/NCT05254847
  • Kuang M Adjuvant immunotherapy combined with chemoradiation for patients with high-risk reseCtable extrahepatic chOlangiocarcinoma and gallblaDder cancer: a phase II, multicenter, randomized controlled trial [Internet].Report No.: NCT04333927. clinicaltrials.gov; 2020 [cited 2024 Jan 1]. Available from: https://clinicaltrials.gov/study/NCT04333927
  • Institut für klinische krebsforschung IKF GmbH at Krankenhaus Nordwest. A phase II study of immunotherapy with durvalumab and tremelimumab in combination with capecitabine or without capecitabine in adjuvant situation for biliary tract cancer [Internet]. Report No.: NCT05239169. clinicaltrials.gov; 2024 [cited 2024 Jan 1]. Available from: https://clinicaltrials.gov/study/NCT05239169
  • Shanghai Zhongshan Hospital. A randomized controlled, multicenter, open-label, phase II clinical study of PD1 antibody (toripalimab) combined with GEMOX Chemotherapy and lenvatinib neoadjuvant treatment for resectable intrahepatic cholangiocarcinoma with high-risk recurrence factors [internet]. Report No.: NCT04506281. clinicaltrials.gov; 2020 [cited 2024 Jan 1]. Available from: https://clinicaltrials.gov/study/NCT04506281
  • Chen W, Xu D, Liu Q, et al. Unraveling the heterogeneity of cholangiocarcinoma and identifying biomarkers and therapeutic strategies with single-cell sequencing technology. Biomed Pharmacother. 2023;162:114697. doi: 10.1016/j.biopha.2023.114697
  • Connor AA, Gallinger S. Pancreatic cancer evolution and heterogeneity: integrating omics and clinical data. Nat Rev Cancer. 2022;22:131–142. doi: 10.1038/s41568-021-00418-1
  • Mody K, Jain P, El-Refai SM, et al. Clinical, genomic, and transcriptomic data profiling of biliary tract cancer reveals subtype-specific immune signatures. JCO Precis Oncol. 2022;6:e2100510. doi: 10.1200/PO.21.00510
  • Parikh AR, Leshchiner I, Elagina L, et al. Liquid versus tissue biopsy for detecting acquired resistance and tumor heterogeneity in gastrointestinal cancers. Nat Med. 2019;25:1415–1421. doi: 10.1038/s41591-019-0561-9
  • Pellecchia S, Viscido G, Franchini M, et al. Predicting drug response from single-cell expression profiles of tumours. BMC Med. 2023;21:476. doi: 10.1186/s12916-023-03182-1
  • Zhang J, Zhang H, Zhang L, et al. Single-Cell transcriptome identifies drug-resistance signature and immunosuppressive microenvironment in metastatic small cell lung cancer. Adv Genet Hoboken NJ. 2022;3:2100060. doi: 10.1002/ggn2.202100060
  • Bueschbell B, Caniceiro AB, Suzano PMS, et al. Network biology and artificial intelligence drive the understanding of the multidrug resistance phenotype in cancer. Drug Resist Updat. 2022;60:100811. doi: 10.1016/j.drup.2022.100811
  • You Y, Lai X, Pan Y, et al. Artificial intelligence in cancer target identification and drug discovery. Signal Transduct Target Ther [Internet]. 2022;7 [cited 2023 Oct 30]. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9090746/
  • Patel SK, George B, Rai V. Artificial intelligence to decode cancer mechanism: beyond patient stratification for precision oncology. Front Pharmacol [Internet]. 2020 [cited 2023 Oct 30];11. doi: 10.3389/fphar.2020.01177
  • Hayashi H, Uemura N, Matsumura K, et al. Recent advances in artificial intelligence for pancreatic ductal adenocarcinoma. World J Gastroenterol. 2021;27:7480–7496. doi: 10.3748/wjg.v27.i43.7480
  • Applying artificial intelligence to big data in hepatopancreatic and biliary surgery: a scoping review. Artif Intell Surg. 2023;3:27–47. doi: 10.20517/ais.2022.39
  • Beatty GL, Werba G, Lyssiotis CA, et al. The biological underpinnings of therapeutic resistance in pancreatic cancer. Genes Dev. 2021;35:940–962. doi: 10.1101/gad.348523.121