Improving the prognosis of pancreatic cancer: insights from epidemiology, genomic alterations, and therapeutic challenges

Zhichen Jiang , Xiaohao Zheng , Min Li , Mingyang Liu

Front. Med. ›› 2023, Vol. 17 ›› Issue (6) : 1135 -1169.

PDF (3106KB)
Front. Med. ›› 2023, Vol. 17 ›› Issue (6) : 1135 -1169. DOI: 10.1007/s11684-023-1050-6
REVIEW

Improving the prognosis of pancreatic cancer: insights from epidemiology, genomic alterations, and therapeutic challenges

Author information +
History +
PDF (3106KB)

Abstract

Pancreatic cancer, notorious for its late diagnosis and aggressive progression, poses a substantial challenge owing to scarce treatment alternatives. This review endeavors to furnish a holistic insight into pancreatic cancer, encompassing its epidemiology, genomic characterization, risk factors, diagnosis, therapeutic strategies, and treatment resistance mechanisms. We delve into identifying risk factors, including genetic predisposition and environmental exposures, and explore recent research advancements in precursor lesions and molecular subtypes of pancreatic cancer. Additionally, we highlight the development and application of multi-omics approaches in pancreatic cancer research and discuss the latest combinations of pancreatic cancer biomarkers and their efficacy. We also dissect the primary mechanisms underlying treatment resistance in this malignancy, illustrating the latest therapeutic options and advancements in the field. Conclusively, we accentuate the urgent demand for more extensive research to enhance the prognosis for pancreatic cancer patients.

Keywords

pancreatic cancer / cancer screening / single cell / molecular alterations / precancerous lesion / therapy resistance

Cite this article

Download citation ▾
Zhichen Jiang, Xiaohao Zheng, Min Li, Mingyang Liu. Improving the prognosis of pancreatic cancer: insights from epidemiology, genomic alterations, and therapeutic challenges. Front. Med., 2023, 17(6): 1135-1169 DOI:10.1007/s11684-023-1050-6

登录浏览全文

4963

注册一个新账户 忘记密码

1 Introduction

Pancreatic cancer is a malignancy with a particularly poor prognosis with a 5-year survival rate of approximately 12%. It is projected to ascend as the second leading cause of cancer-related mortality in the United States by 2040 [1]. The advanced clinical stage prior to surgery and elevated postoperative recurrence rate critically impede the long-term survival prospects for pancreatic cancer patients [2]. Only 20% of patients have the opportunity for pancreatectomy, while 50% of patients have already developed distant metastases [3,4]. Pancreatic cancer is prone to develop resistance to chemotherapy and immune therapy, which limits the long-term survival of cancer patients. In recent years, numerous multi-omics studies have been conducted, which enriches our understanding of pancreatic cancer microenvironment and precancerous lesions. This article aims to provide a comprehensive overview of various aspects of pancreatic cancer, including its epidemiology, etiology, treatment approaches, diagnosis methods, and mechanisms of drug resistance, as well as an exploration of this disease’s genomic and clinical features.

2 Pancreatic cancer pathogenesis epidemiology, etiology, to mechanism

2.1 Epidemiology

In 2018, global statistics indicated 458 918 new instances of pancreatic cancer, resulting in 432 242 fatalities [5]. The majority of pancreatic cancer cases are pancreatic ductal adenocarcinoma (> 90%). The disease manifests slightly more frequently in males than females, with a ratio of 1.4 to 1.0. Metastases often involve organs such as the liver, lymph nodes, lungs, and peritoneum [4]. The low 5-year survival rate of pancreatic cancer is largely attributed to the advanced stage at diagnosis. Only about 20% of patients present with early and surgically resectable disease, while 50% have metastatic disease and the remaining 30% have locally advanced disease with extensive vascular involvement [2]. Post-surgical patients witness a 5-year survival rate hovering around 15%–25%. Incidences of pancreatic cancer are rare below 40 years of age, yet both its prevalence and mortality rates escalate with advancing age [68]. Additionally, the incidence of pancreatic cancer is significantly higher in high-income countries compared to those with a median or low human development index, which is thought to be associated with lifestyle factors [5].

Routine screening for pancreatic cancer in high-risk groups is not recommended as standard practice. Research by Canto and colleagues revealed that around 7% of those deemed at heightened risk for pancreatic cancer received a diagnosis within a 16-year span. The median interval between initial screening and eventual diagnosis stood at 4.8 years (with an interquartile range of 1.6–6.9 years). Prolonged monitoring of individuals susceptible to pancreatic cancer showed that most malignant growths identified during these check-ups were operable, consequently improving the outlook [9]. However, Corral et al. demonstrated that in routine endoscopic screening for high-risk individuals, it is estimated that only after screening 135 high-risk patients, can one patient be identified as having high-risk pancreatic lesions [10]. Consequently, additional research is required to identify more efficient screening indicators that can be advantageous for high-risk populations in terms of cancer detection.

2.2 Etiology

2.2.1 Short-term factors

Short-term risk factors for pancreatic ductal adenocarcinoma harbor a notably higher degree of peril compared to their long-term counterparts, primarily as they frequently emerge as harbingers of disease progression. Manifestations such as cachexia, the onset of diabetes, and the advent of pancreatitis could all be clandestine indicators of pancreatic cancer. Both weight loss and cachexia could serve as premonitory signals of precancerous stages in pancreatic malignancy, with the ensuing risk escalating in tandem with the degree of weight loss [11]. New-onset diabetes may also suggest the presence of underlying pancreatic cancer. Patients newly diagnosed with diabetes exhibit a 0.3% to 1% likelihood of developing pancreatic cancer within the initial three years following their diabetes confirmation, a rate that notably surpasses that of individuals without diabetes [1214]. Similarly, pancreatitis may be a consequence of underlying pancreatic cancer, resulting in a significantly higher risk of new-onset pancreatitis within the first year compared to a long-term diagnosis [15].

2.2.2 Long-term factors

Resource from the World Health Organization (WHO) Global Health Observatory database shows that smoking, alcohol drinking, physical inactivity, obesity, diabetes, hypertension, and high cholesterol are long-term risk factors for pancreatic cancer after age-adjustment [16]. Smoking has been identified as the predominant lifestyle risk factor associated with pancreatic cancer. A comprehensive meta-analysis involving 12 case-control studies, with a cohort of 6507 pancreatic cancer patients and 12 890 controls, revealed an odds ratio (OR) of 1.74 (95% confidence interval (CI) 1.61–1.87) demonstrating the correlation between smoking and pancreatic cancer [17,18]. People who smoke more than 35 cigarettes a day have a higher risk of developing pancreatic cancer compared to never-smokers [17]. Cessation of smoking diminishes this risk. Notably, ex-smokers witness a progressive decline in pancreatic cancer risk correlating with the duration since they stopped smoking, yet their risk remains substantially elevated compared to individuals who have never smoked, particularly within the first decade. However, the risk level reverts to that akin to never-smokers after a span of 10–20 years post-quitting [7,18].

Diabetes may be factors that increase the prevalence of pancreatic cancer in younger patients [19]. The long-term risk (> 3 years) of pancreatic cancer increases with the time to diagnosis of diabetes but the short-term risk (< 3 years) decreases as the duration of diabetes diagnosis increases [20,21]. Beyond the mere presence of a diabetes diagnosis, specific diabetes-related biomarkers, such as levels of glucose and insulin, are also linked to an elevated risk of developing pancreatic cancer [7,8].

Obesity [19] and high alcohol intake [22] correlate with a heightened risk ratio, mirroring the extent of their severity. Particularly, excessive intake of alcohol is connected to the onset of pancreatitis, a known precursor for pancreatic cancer [23]. Patients with chronic pancreatitis are at a higher risk of pancreatic cancer owing to persistent inflammation and injury. In a pooled analysis of 14 prospective cohort studies of 862 664 individuals, chronic pancreatitis was associated with a 13.3-fold increased relative risk of pancreatic cancer (95% CI 6.1–28.9) [22].

We found that there is a clear association between the above common long-term risk factors and the demographics of pancreatic cancer. The prevalence of pancreatic cancer in the population was associated with population of smoking, diabetes, obesity and with secondary pancreatitis owing to heavy alcohol consumption, demonstrating the chances of controlling risk factors to reduce the incidence of pancreatic cancer [6,23].

2.2.3 Environmental factors

Numerous studies delving into the interplay between environmental determinants and the risk of pancreatic cancer, encompassing aspects such as viral infections, occupational exposures, and microbiological constituents, have often yielded incongruent results. Virus infection may be associated with an increased risk of pancreatic cancer. The conclusions about whether hepatitis B and hepatitis C increase the risk of pancreatic cancer are contradictory. Although a meta-analysis of eight studies reported that both hepatitis B and C infection were associated with an increased risk of pancreatic cancer [24], other studies have provided inconsistent conclusions [2527]. There was no overall association between Helicobacter pylori infection and the risk of pancreatic cancer, but some subtypes of H. pylori strains may have an impact [28]. Patients are occupationally exposed to aromatic hydrocarbon solvents, pesticides, chlorinated hydrocarbon solvents, formaldehyde, volatile sulfur compounds, and heavy metal exposure with a higher risk of developing pancreatic cancer [29]. Zaitsu et al. supported occupational class differences affecting the survival of pancreatic cancer. The service, blue-collar, and unemployed individuals had significantly lower survival rates than white-collar workers [30]. Rare subgroups of oral microbes can also increase pancreatic risk [31]. However, a history of allergy is a protective factor against pancreatic cancer, including a history of hay fever and animal allergy, chronic asthma, and nasal allergy [32].

2.2.4 Inherited factors

2.2.4.1 Family history of pancreatic cancer

Familial instances of pancreatic cancer may result from a combination of common environmental exposures and inherent genetic predispositions. Individuals with a familial background of pancreatic cancer exhibit a more frequent occurrence of conditions like pancreatic intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasia (IPMN) within their healthy pancreatic tissue, in contrast to those lacking such family medical history. The risk of pancreatic cancer is also elevated in families with relatives diagnosed with other cancers [4]. However, patients with or without a family history of pancreatic cancer develop the disease in a very similar manner. There were no statistically significant differences in pathological features and somatic mutation profiles between familial and sporadic pancreatic cancer cases [33]. The long-term follow-up results of pancreatic cancer families have also revealed that the risk of developing pancreatic cancer in individuals who do not carry key mutations is close to 0, significantly lower than the 9.3% risk observed in those with pancreatic cancer susceptibility gene mutated (CDKN2A, LKB1/STK11, BRCA1, BRCA2, PALB2, TP53, MLH1, MSH2, MSH6, ATM) [34]. These findings suggest that the genetic basis of familial and sporadic pancreatic cancer might be highly similar.

2.2.4.2 Genetic susceptibility genes

Multiple genome-wide association studies (GWAS) have pinpointed critically pathogenic germline mutation loci integral to the genesis of pancreatic cancer. Patients harboring germline single nucleotide polymorphisms (SNPs) confront a substantially escalated lifetime risk of pancreatic cancer juxtaposed against their non-mutated counterparts [35]. Germline mutations occur in 4%–10% of pancreatic cancer patients, particularly in those with DNA repair-related mutations, such as homologous recombination defects and Lynch syndrome [36,37]. The BRCA2 pathogenic variant is the most common high-risk genetic variant found in pancreatic cancer patients, accounting for 2%–17% of cases. Individuals with pathogenic BRCA2 variants have a 3.5–5.8-fold increased risk of developing pancreatic cancer compared to those without these variants [38]. Other genes associated with pancreatic cancer risk include PALB2 (< 0.5%), BRCA1 (0.6%–2.2%), ATM (2.3%), STK11 (< 1%), P16/CDKN2A (< 1%–2.5%), PRSS1 (< 1%), MLH1 (< 1%), MSH2 (< 1%), MSH6 (< 1%), PMS2 (< 1%), and various other germline mutations commonly found in syndromes and tumors [39]. Genetic counseling is recommended for both patients and healthy family members who meet certain criteria, such as having a first-degree relative with early-onset pancreatic cancer (< 50 years old), more than one first-degree family member with pancreatic cancer, or a known pathogenic germline gene mutated associated with pancreatic cancer [4,40].

2.3 Mechanism

2.3.1 Somatic mutations

The predominant somatic gene alterations in pancreatic ductal carcinoma include KRAS mutations (90%), TP53 mutations (50%–74%). Approximately 90% of all grades of pancreatic cancer have activated oncogenic KRAS mutations. G12D, G12V, and G12C mutations are the most common, while G12R and G12A mutations, as well as other point mutations at codons 11, 13, 61, or 146, appear to be less common [41,42]. KRAS, a membrane-associated GTPase, mediates cellular growth signals via the MAPK and PI3K pathways. Predominant oncogenic mutations in KRAS are known to disturb the equilibrium of KRAS GTP–GDP cycling [41]. Meanwhile, the deactivation of the tumor suppressor gene TP53 hinders the identification of DNA damage and prevents cell cycle halt, enabling cells to ignore cell cycle checkpoints and resist apoptotic cues. Other mutations are less common, including alternative driver variants in non-KRAS mutant pancreatic cancer, low-frequency mutations associated with precancerous lesions, and critical pathway adaptation mutations.

Remarkably, approximately 10% of pancreatic cancers are devoid of activating KRAS mutations and demonstrate superior outcomes relative to their KRAS-mutated counterparts [41]. These cases are notable for mutations or copy number alterations in alternative drivers such as activating mutations or amplifications of BRAF, FGFR1, or ERBB2, inactivating mutations in NF1, DUSP6, or SPRED1 [43,44] or fusions involving NRG1 and NTRK1 [4547], alternative oncogenic events genes in SWI/SNF and COMPASS complexes [48], and other familial syndrome genes [43]. Mutations in GNAS are only observed in IPMN with only about 4% of pancreatic cancer patients because IPMN is not a major evolutionary precursor in pancreatic cancer [49]. Most of these low-frequency mutations occur in genes related to cellular processes such as cell survival, cell fate determination, and genomic maintenance [43].

2.3.2 Chromosome alterations

Pancreatic cancer develops complex copy number variants (CNVs) throughout the genome during progression, which are categorized as arm-level and focal-level CNV. Common arm-level alterations include amplifications of 1q (33%) and deletions of 6p (41%), 6q (51%), 8p (28%), 9p (48%), 17p (64%), 17q (31%), 18p (32%), and 18q (71%), which result in significant expression changes in genes, proteins, and phosphoproteins [50,51]. Repeated focal CNVs encompass recognized oncogenic propellants, featuring amplifications in GATA6 (18q11.2), ERBB2 (17q12), KRAS (12p12.1), AKT2 (19q13), and MYC (8q24.2), in addition to deletions in CDKN2A (9p21.3), SMAD4 (18q21.2), ARID1A (1p36.11), and PTEN (10q23.31) [43,52]. Deletions of CDKN2A and SMAD4 are most commonly observed alternations. Alterations in CDKN2A are also early events, while alterations of SMAD4 are late events. Approximately 31%–38% of pancreatic cancer has SMAD4 mutations reducing SMAD4-dependent TGF-β inhibition and promotes non-canonical TGF-β signaling, thereby resulting in reduced cell cycle arrest and apoptosis, promotion of epithelial-mesenchymal transition (EMT) and angiogenesis, and induction of immune suppression, all of which contribute to the progression and metastasis of cancer cells [3,53,54]. Nonetheless, research employing genetically modified mice indicates that a pancreatic-specific lack of SMAD4 does not trigger PanIN or invasive pancreatic cancer [48]. Alterations in the copy number of GATA6 and MYC partially characterize the pancreatic cancer phenotype [48]. Severe imbalance of KRAS and tetraploidization is also more often observed CNV in clinically advanced or metastatic samples, which are critical for molecular subtyping [55].

A key factor in the onset and development of pancreatic ductal adenocarcinoma involves the epigenetic changes impacting oncogenes and tumor suppressor genes. Such alterations, which are reversible, transform the structure of chromatin and histones, thereby modifying the accessibility of gene promoters and the patterns of gene expression. In the context of pancreatic tumor development, histone methylation and acetylation stand out as the most crucial forms of histone adjustments [56]. DNA methyltransferase, including DNMT1, DNMT3A, and DNMT3B, have been shown to be increased in pancreatic cancer and are associated with lower overall survival [57,58]. Methylation of DNA in the promoter regions of tumor suppressor genes like APC, BRCA1, and CDKN2A impedes their transcriptional activities. This phenomenon is considered to be linked to the pathogenesis of human pancreatic cancers [56].

DNA methyltransferase, including DNMT1, DNMT3A, and DNMT3B, have been shown to be increased in pancreatic cancer and are associated with lower overall survival [57,58]. DNA methylation of tumor suppressor genes APC, BRCA1, and CDKN2A at their promoter regions blocks transcription activity, which is thought to be associated with human pancreatic cancers [56].

2.3.3 Spatial transcriptomics

Spatial omics is a well-established concept, with RNAscope and multiplex immunohistochemistry (mIHC) being capable of achieving single-cell resolution for a long time. However, the high cost has limited the widespread use of high throughput methods. In recent years, there has been a gradual increase in the throughput of spatial technologies. Currently, high-throughput spatial technologies can be broadly classified into fluorescence in situ hybridization (FISH)-based spatial transcriptomics, sequencing-based spatial transcriptomics, multiplexed imaging based-proteomics, and spatial mass spectrometry-based proteomics. The clinical and translational values of spatial omics in pancreatic cancer have been well elucidated (Tab.1) [59]. Current spatial transcriptomics techniques are limited by their throughput and resolution, because the gene expression detected by spatial spots is a mixture [60]. Spatial proteomics has also been used for pancreatic cancer; however, the number of detected genes has been limited (< 50) compared to spatial transcriptomics [61,62].

Moncada et al. published the first protocol using the Multimodal Intersection Analysis (MIA) algorithm for single-cell sequencing and spatial transcriptomics mapping. They found that macrophages were most enriched immune cell type in pancreatic cancer. Moreover, specific subgroups of ductal cells, macrophages, dendritic cells (DCs), and various other cell types show a spatially confined enrichment in certain compartments. Particularly, M1 macrophages are predominantly found in stromal areas as well as cancerous tissues, indicating the presence of an inflammatory milieu in these zones [63]. Hwang et al. [64] employed NanoString GeoMx DSP to unravel the spatial arrangement of cells and expression patterns within multicellular structures. Their findings highlighted that the majority of malignant programs demonstrated greater variability between individual patient tumors than among different regions of interest (ROIs) within a single tumor. However, mesenchymal, immunomodulatory, and myofibroblastic progenitor programs were relatively more stable. Notably, the neural-like progenitor and neuroendocrine-like malignant programs were more prevalent in ROIs derived from tumors treated with chemoradiotherapy (CRT) compared to those untreated, aligning with insights from single-nucleus RNA sequencing (snRNA-seq) analyses. These spatial relationships encompass both extensive multicellular communities, evident from clustering analysis, and more nuanced features seen in specific association pairs. Zhou et al. [65] used a spatial sequencing platform to find the spatial distribution of pancreatic cancer with different driver mutations status. Validating acinar-to-ductal metaplasia (ADM) using spatial transcriptomics data remains challenging due to the lack of single-cell resolution and the scarcity of ADM cells. Nonetheless, regions containing ADM cells might exhibit expression markers characteristic of both acinar and ductal cells, a phenomenon that aligns with the findings presented by Tosti et al. [66]. Zhou et al. identified NECTIN4 as the NECTIN most specifically associated with tumor cells. Through spatial transcriptomics data, our analysis concentrated on two particular regions to examine the expression of TIGIT in areas close to regions infiltrated by lymphocytes. This investigation revealed a colocalization of tumor regions in the H&E stains with NECTIN4 expression, underscoring the potential of the TIGIT–NECTIN4 axis as a therapeutic target to enhance anti-tumor T cell activity [65]. Barkley et al. [67] utilized spatial data to demonstrate the induction of a squamous module in pancreatic cancer, pointing to a process of squamous differentiation. This includes differentiating between classical subtypes (characterized by elevated expression of acinar-ductal genes like TFF1 and CEACAM6) and basal subtypes (marked by higher levels of squamous and basal genes, such as LY6D and KRT15). The heightened expression of squamous markers in pancreatic cancer implies a frequent partial metaplasia, steering toward a squamous program [68]. Progressive pancreatic cancer expresses more squamous-associated genes compared to normal pancreas, and squamous staging has been associated with poorer prognosis. It is important to note that changes in gene expression do not imply pathomorphologic conversion of adenocarcinoma to squamous or adenosquamous carcinoma, which are rare in histology [69]. Spatial results were also used to visualize hypoxic microenvironment-induced spatial transcriptome changes in pancreatic cancer [70]. Bell et al. applied migration learning to integrated PanIN spatial transcriptomics with single-cell RNA sequencing (scRNA-seq) data, allowing analysis of cellular and molecular progression from PanIN to pancreatic cancer. Using PanIN samples, it was found that PanIN lesions are predominant in the classical pancreatic cancer subtype. For the first time, it was observed that the same cancer-associated fibroblast (CAF) subtype (myofibroblasts (myCAF), inflammatory CAF (iCAF), and antigen-presenting CAF (apCAF)) present in aggressive pancreatic cancer is also present in human precancerous lesions. CAF-PanIN interaction promotes inflammatory signaling in tumor cells. As PanIN progresses to pancreatic cancer, tumor cells shift to proliferative signaling [71]. Two other unpublished studies identified NKX6-2 as a key transcription factor for the fatal transition in IPMN using spatial transcriptomics [72,73].

2.3.4 Single cell screening

In the past, we used bulk RNA sequencing (RNA-seq) results using paired tumor and normal specimens, and the sequencing results are mixture of each cell. The single cell era has greatly enriched our understanding of tumor microenvironment (TME) of pancreatic cancer, which provides us further insights of tumor and TME cells.

2.3.4.1 Untreated pancreatic cancer cells

Peng et al. [74] classified untreated pancreatic cancer separated into type 1 and 2 ductal cells using single cell RNA sequencing results. Type 1 ductal cells are relative normal and present in both non-cancerous and neoplastic tissues, which were enriched for cell adhesion, migration, and inflammatory response, while type 2 ductal cells were enriched for malignant phenotypes such as proliferation and hypoxia. Repair of the damaged pancreas generates ADM, which is essential for recovery. Although this repair process is reversible, post-repair dedifferentiation can be prevented by the introduction of KRAS mutations and recurrent inflammation, which leads to the formation of PanIN, a precursor condition for pancreatic cancer. In addition, concomitant deletion of tumor suppressors such as p53 can directly lead to pancreatic cancer initiation, and tuft cells can also convert to PanIN after mutation [75].

Surgically resected primary tumor is different from sampling results. Because metastatic puncture samples from recurrent metastases have a higher proportion of epithelial cells compared to recent scRNA-seq studies of surgically resected pancreatic cancer tumors, the most common immune cell types are myeloid and T cells, both monocytes and the higher proportion of invasive basal-like cell subtypes found in metastases by bulk RNA-seq are more likely to be present [49,76,77].

2.3.4.2 Fibroblasts

Fibroblasts are key cells in the mesenchymal environment of pancreatic cancer, and fibroblasts have pro-tumor proliferation and EMT effects [78]. Both iCAF and myCAF can be conservatively identified in most cancers. iCAF is characterized by ACTA2low FAPhigh and myCAF is characterized by ACTA2high and most of the single cell studies require that fibroblasts in pancreatic cancer have ACTA2 expression. These two fibroblast populations are the most common fibroblast subpopulations in all prior tumors and can be conservatively identified in multiple single-cell sequencing. Most of the cell subpopulations in single-cell studies of pancreatic cancer can be classified into these two groups. apCAF is further suggested by the three CAF isoforms (myCAF, iCAF, and apCAF) described by Elyada et al. apCAF subpopulation is defined as CD74+HLA+CD45 fibroblasts, however, this idea remains clearly controversial [79]. apCAF is sometimes difficult to be conservatively identified. In particular, apCAF has a very low percentage by itself and HLA+CD74+ is widely expressed between cells. The CAF derived from murine pancreatic cancer forms a separate cluster, whereas apCAF detected in human pancreatic cancer is scattered in the iCAF and apCAF clusters. A number of pancreatic cancer single-cell data set analyses have failed to identify apCAF, although they do not deny this possibility. One thing is confirmed, apparently in human pancreatic cancer single-cell data sets, apCAF is not able to form an independent group like iCAF and myCAF [64,76]. Another paper by Zhou et al. not only identified apCAF, myCAF, iCAF, but also CD133high iCAF and CXCR4high iCAF, CD133high iCAF expressing both stem cells and the epithelial marker EPCAM, and CXCR4high iCAF expressing strongly CD45. These two categories are like epithelial and immune cells, and we still need to look at them with caution [65]. Fibroblast cells are conserved across organs. Since there is no gold standard for fibroblasts, there are a variety of typologies, such as steady-state-like (SSL), mechanoresponsive (MR), and immunomodulatory (IM) CAFs [80]. scRNA-seq reveals stromal evolution into LRRC15+ myCAF as a determinant of patient response to cancer immunotherapy. Dominguez found PDPN+ cells are the dominant fibroblast population in normal and pancreatic cancer. Levels of the LRRC15+ CAF were correlated with poor response to anti-PD-L1 therapy. Late-stage tumors support previous observations identifying IL-1-driven iCAF and TGF-β-driven myCAF [81]. Such results explain the complex crosstalk between fibroblast heterogeneity and cancer immunity [8284].

2.3.4.3 Immune cells

Steele et al. compare the difference of immune cells from blood to pancreatic tissue [85]. Multiomic profiling of lung and liver tumor microenvironments of metastatic pancreatic cancer reveals site-specific immune regulatory pathways, TME of lung generally exhibits higher levels of immune infiltration, immune activation, and pro-immune signaling pathways in lung cancer, whereas multiple immune-suppressive pathways are emphasized in the liver TME [86]. Within the myeloid cluster, subclustering revealed six distinct populations that were identified as resident macrophages, alternatively activated M2-like macrophages, classic monocytes, cDC1, and two types of Langerhans-like DC. Within T cell cluster, subclustering identified four discrete cell types: CD8+ T cells, CD4+ T cells, regulatory T cells (Tregs), proliferating T cells. In addition, several groups have examined features of the tumor immune microenvironment during the tumorigenic process, but suitable immune checkpoint blockade therapy in pancreatic cancer is still lacking [87].

2.3.4.4 Treated pancreatic cancer

scRNA-seq reveals the effects of chemotherapy on human pancreatic adenocarcinoma and its tumor microenvironment. Different iCAF were observed under different chemotherapy analyses. Naïve patients are both low in two lists of genes, with HSPA1A1, HSPA1AB, DNAJB1, FOS, JUN, FOSB high after FOLFIRINOX (5-FU/calcium folic plus oxaliplatin and irinotecan), and MT1X, MT1M, MT1E, and MT2A high after gemcitabine (GEM) plus nab-paclitaxel [65]. Hwang and his colleagues, Shiau et al., presented a single-cell resolution framework of diverse remodel of states and distributions in endothelial cells enriched with vasculogenesis, stem-like state, response to wounding and hypoxia, and endothelial-to-mesenchymal transition (reactive EndMT). A bulk transcriptome analysis of two independent cohorts (n = 269 patients) revealed that the lymphatic and reactive EndMT lineage programs were significantly associated with poor clinical outcomes [88].

2.3.5 High-throughput sequencing application

Other high-throughput sequencing technologies are also widely used in the study of pancreatic cancer, including high-throughput/resolution chromosome conformation capture (HI-C) sequencing technology, the third generation DNA/RNA sequencing technology, cleavage under targets and release using nuclease (CUT&RUN), methylation sequencing technology, epigenomics, and other technologies. Although these technologies are characterized by having very good sequencing results of cancer cell lines, results in pancreatic tissues need to be interpreted with caution, due to the low tumor purity of pancreatic cancer.

(1) Long-read sequencing: Du et al. discovered structural variants as well as chromatin structure of pancreatic cancer using Hi-C and long-read sequencing. Structural variants are defined as variants more than 50 bp, and structural variants located in key genetic regions can also drive tumor malignant progression [89].

(2) scATAC-seq: Single-cell assay for transposase-accessible chromatin using sequencing (scATAC-seq) and single-cell combinatorial indexing ATAC-seq (sci-ATAC-seq) are effective tools in the study of genome-wide chromatin accessibility landscapes. Chromatin region accessibility play essential roles in epigenetic regulation have been measured at the single-cell level using single-cell ATAC-seq approaches [90,91].

(3) scCOOL-seq: scCOOL-seq is a single-cell multi-omics sequencing technology that can analyze DNA methylation and chromatin accessibility togather. Fan et al. found ZNF667 and ZNF667-AS1, whose expressions were linked to a better prognosis for pancreatic cancer patients by affecting the proliferation of cancer cells and revealed the critical epigenomic features of cancer cells in pancreatic cancer patients at the single-cell level [92,93].

2.4 Molecular subtypes

Waddell et al. used the results of high-throughput DNA sequencing to establish the first molecular typing of pancreatic cancer based on genomic variants, and classified pancreatic cancer into stable, locally rearranged, scattered and unstable [94]. The molecular typing of pancreatic cancer was established for the first time based on genetic and genomic variant sequencing. In 2011, Collisson et al. reported three molecular subtypes of pancreatic cancer based on the analysis of 27 microdissected pancreatic cancer samples: classical, quasimesenchymal (QM-PDA), and exocrine-like. In 2015, Moffitt et al. [95] clustered two states of pancreatic ductal adenocarcinoma epithelium into 2 types: classical and basal. The classical types of high GATA6 expression and the basal subtypes have stronger EMT, and the basal types have more squamous cell component expression, and worse prognoses. In 2016, Bailey et al. [69] used unsupervised clustering of RNA-seq data from 96 tumors with at least 40% epithelial content and yielded four subtypes: squamous, pancreatic progenitor, immunogenic, and abnormally differentiated endocrine exocrine (ADEX). Raphael et al. [43] found that reconciliation of previously identified subtypes and high-purity tumors can consistently be classified into either the basal-like/squamous or the classical/progenitor subtype. Although no entirely new typing was introduced, it was suggested that the results of three major methods of molecular classifications were heavily influenced by tumor purity, and that high-purity samples conformed to the classical-basal class, establishing the Moffit typing in pancreatic ductal cell carcinoma. In 2018, Puleo et al. [96] found the exocrine-like (Collisson et al. [97]) and ADEX (Bailey et al.) subtypes resulting from a mixture of pancreatic endocrine cells, acinar cell contamination, or acinar cell carcinoma, and refined the subtypes into activated, desmoplastic, pure classical, and immune subtypes. Given that Moffit has become the most important subtype reflecting pancreatic ductal cell carcinoma, it is limited by the software calculations responsible and inconsistencies in tumor purity. Significant differences appeared in the tissue composition of pancreatic tumors in the Moffitt, Bailey, and TCGA cohorts. Up to 45% of tumor samples from various public cohorts were reclassified after removal of non-epithelial signals from tumor bulk expression profiles [98]. Indeed, there was significant sampling heterogeneity across tumor sites and it is inconclusive whether this sampling error affects the correct therapeutic staging of patients [99]. In 2020, Rashid et al. create the benchmark classifier Purity Independent Subtyping of Tumors (PurIST) to allow clinicians to better reproduce Moffitt subtypes for all samples in clinical trials in bulk RNA-seq [100]. PurIST found a purity-independent subtypes of classical and basal-like subtypes, using the NanoString platform and the Illumina sequencing platforms for bulk RNA-seq results. Moffitt typing of pancreatic cancer RNA-seq results on different platforms can be performed with over 95% accuracy based on a simple formula without any fiber-cutting techniques, however, unfortunately this tool cannot be used for single cell sequencing results. In 2022, CPTAC–pancreatic ductal adenocarcinoma (PDAC) started the first addition of proteomics multimodal typing, using multi-omics NMF typing to classify pancreatic cancer into 4 subtypes, and found that the typing is highly influenced by sampling location and purity. When retaining high purity samples of pancreatic cancer, the samples can be two classes, the typing with Moffitt dichotomy including Cluster 1 (Moffitt classical) and Cluster 2 (Moffitt basal), which is highly correlated with prognosis. It is important to note that protein-based dichotomization or RNA dichotomization is not always consistent. xCELL typing was used by CPTAC–PDAC to determine immune hot-cold subtypes and found that 92.3% samples (108 samples/117 samples) had little or no immune infiltration. Only 7.7% of the samples were from immune hot tumors, and immune hot tumors had significantly lower tumor purity. If 40 samples rich in follicular and islet cells were removed (probably due to essential tumor characteristics or early tumor staging or insufficient tumor composition at the location of harvest), 88.5% (69 samples/78 samples) patients were definite immune cold tumors. 11.5% of the visualized sections were examined after removing false positives at the sampling site due to mixed lymph nodes. The remaining 88.5% (69 samples/78 samples) had definite immune cold tumors [50]. These findings support that pancreatic cancer is a naturally immunotherapy-resistant tumor with little clinical response to PD-1/PD-L1 blockade therapy. The concomitant endothelial cell remodeling characterized by decreased expression of endothelial adhesion proteins, accompanied by elevated VEGF and hypoxic pathways, increased glycolysis and dysregulation of cellular junctions in immune-cold pancreatic cancer may form a common inhibitor of immune cell infiltration. Adapting pancreatic cancer to immune hot tumors will be the most important target in the future [101103]. The most important contribution of CPTAC–PDAC is the validation limiting the previous staging of immune prototype tumors to a very small fraction of pancreatic cancers and the need to exclude false positives.

In 2020, COMPASS/PanCuRx began to explore molecular subtypes using both bulk RNA-seq and scRNA-seq, and they classified pancreatic cancer into 5 groups: basal-like A, basal-like B, hybrid, classical A, classical B [55]. Similar to the coexistence of basal and classical programs within the tumor, this mixture of cell states may be more consistent with the true response of tumor cell heterogeneity. Patients with basal-like A pancreatic cancer usually present with advanced disease and have the worst response to (GEM)-based chemotherapy and FOLFIRINOX. In contrast, patients with basal-like B and mixed tumors usually present with resectable disease. Thus, the ability to distinguish basal-like A, basal-like B, and hybrid subtypes from groups previously classified as basal-like allows for more accurate prediction of chemotherapy response. Classical A/B tumors were found to be associated with increased frequency of GATA6 amplification and complete loss of SMAD4, whereas basal-like A/B tumors showed complete loss of CDKN2A and higher frequency of TP53 mutations. At single-cell resolution, results of COMPASS/PanCuRx also show that basal-like and classical subtypes can coexist in the same tumor, highlighting the molecular heterogeneity within the tumor. In fact, many subtypes are not absolutely mutually exclusive and it is more like determining which subtype is characterized by a dominant feature in the overall development as the subtype to which the patient belongs [104]. The patient’s strain is determined by the characteristics of the patient. A change in the dominant cell population may occur after patients underwent pradiation, chemotherapy, or immunotherapy. The use of FOLFIRINOX or GEM plus nab-paclitaxel presented different expression profiles of pancreatic cancer [65]. In the era of single cell, in addition to emphasizing the qualities of the cells themselves, the concept of cell state began to be introduced, where changing the surrounding environment differently exerts a completely different effect on the tumor cells, an effect that can be traced back to specific cell types [64,105]. Raghavan et al. stated that the EMT program is positively correlated with basal-like features and negatively correlated with classical features possessing more basal-like typing in metastatic samples. This may be related to the copy number imbalance of KRAS. Single cell-based typing classified the pancreatic cancer into scBasal, intermediate co-expressor (IC), and scClassical subtypes, using single cell results [105]. An updated molecular subclassification of pancreatic cancer was reported by Hwang et al. in 2022. Using snRNA-seq and spatial transcriptome analysis of 43 major pancreatic cancer samples (18 untreated and 25 treated), they identified three distinct subtypes: classical, squamous-basal, and treatment-enriched. Their study found that the NRP malignant cell program is enriched for residual cancer after radiotherapy. NRP cells are associated with pancreatic cancer through regulation of drug efflux-related genes, negative regulation of cell death, chemoresistance (e.g., ABCB1, BCL2, PDGFD, and SPP1), tumor-neural crosstalk (e.g. SEMA3E, RELN, and SEMA5A) and metastasis (NFIB) of patients with treatment resistance and low survival rates. We identified 14 malignant cell programs that reflected either lineage (classical, squamoid, basaloid, mesenchymal, acinar-like, neuroendocrine-like, and NRP) or cell state (cycling-S, cycling-G2/M, MYC, interferon, tumor necrosis factor/nuclear factor κB (TNF-NFκB), ribosomal and adhesive pathways) and four CAF programs (myofibroblastic progenitor, neurotropic, immunomodulatory, and adhesive). These molecular classifications of pancreatic cancer provide a rich and comprehensive data set to better understand pancreatic tumorigenesis, genetic/molecular landscape, intra- and inter-tumor heterogeneity, tumor progression, and drug resistance. More importantly, molecular subtyping of pancreatic cancer may provide useful information for more effective subtype-tailored treatment of pancreatic cancer patients. However, due to their complexity, these classifications of pancreatic cancer have not yet been used in routine pathological diagnosis or clinical practice (Tab.2).

2.5 ADM

ADM is thought to be the primary origin of pancreatic precancerous lesions that eventually develop into pancreatic ductal adenocarcinoma. The origin of pancreatic ductal adenocarcinoma includes: (1) conversion of acinar cells to PanIN via ductalization and eventually to pancreatic cancer; (2) conversion of acinar cells to other receptor cell types such as tuft cells, and mutation of susceptible cells to malignant cells; (3) direct carcinogenesis of ductal cells; (4) abnormal transformation of pancreatic progenitor cells into tumors; (5) formation of pancreatic cancer through other precancerous lesions secondary to cancer. The ADM origin theory is the most mainstream theory because it can explain the sharp decrease in the number of normal cellular follicular cells, endocrine cells in pancreatic cancer tissue sections. Acinar cells are highly plastic and have the potential to transform into ductal epithelium or endocrine epithelium [66,106]. ADM may be a decisive step in the tumorigenic process, selecting plastic cells for more aggressive subsequent tumorigenesis by transforming acinar cells into malignant cells and precursor fine cells. The formation of ADM was found to be regulated by two major signaling pathways downstream of KRAS (PI3K/Akt and MEK/Erk). Among them, RAC1 is an important target downstream of PI3K/Akt signaling that mediates actin polymerization to redistribute filamentous actin from the apical to the basal part of the acinar cell, leading to apico-basal mechanical tension imbalance. The ADM process in KRAS mutant mice is irreversible and progresses to PanIN, which eventually develops into pancreatic cancer. The pancreatic intraepithelial neoplasia is a unique inflammatory microenvironment-induced pathway of acinar–ADM/PanIN–pancreatic cancer evolution [107]. Two published landscape of combined single-cell and spatial-omics analyses for pancreatic cancer have established temporal model based on the acinar–ADM–PanIN/atypical duct–pancreatic cancer hypothesis [64,65]. Although the progressive model occupies residency, the question of the sequence of ADM and PanIN or whether they must undergo the abovementioned progressive process is not conclusive [108,109].

ADM is defined by conversion of pancreatic acinar cells to abnormal ductal epithelial adenocarcinoma. Using a mouse model where activation of KRAS G12D and deletion of Fbw7 or p53 in acinar cells leads to ADM, Krt19-positive globular lesions with a continuous ductal tree can be observed [107]. It has also been theorized that ADM is a physiological, rapid, and reversible adaptive response capable of limiting the adverse effects of persistent stimuli such as repetitive pancreatitis, with rapid dedifferentiation of acinar cells, thereby producing an effective response by rapidly reducing acinar zymogens (such as amylase, elastase, and pancreatic ribonuclease) to effectively limit tissue damage and enable rapid and complete recovery from it. This adaptation is able to rapidly reactivate ADM during subsequent inflammatory events. Some ADM cells acquire KRAS mutations during this process to adapt to the environment [110].

In contrast to tumor cells, the ADM population expresses oncogenes and significantly upregulates epithelial-mesenchymal transition and stem cell genes. The unique expression pattern of ADM as an intermediate state suggests a dynamic shift between tumor and follicular fate and a role in initiating pancreatic tumorigenesis by gaining access to drive the progression of KRAS events to pancreatic cancer [111]. Single cells make it possible to identify the transition state ADM, which Zhou et al. refer to as ADM_Normal for expression similar to that between the follicular cells and the normal ductal lineage, and ADM_Tumor for transition cells between the follicle and PanIN. While both ADM_Tumor and ADM_Normal showed a decline in the expression of follicular markers, they conversely exhibited an uptick in the expression of pancreatic cancer indicators and catheter-associated markers, respectively. A small fraction of ADM_Tumor cells displayed this trend, indicating that ADM_Normal may represent a transitional phase more akin to regular ductal cells, largely devoid of genomic changes. In contrast, ADM_Tumor appears to be more comparable to PanIN, carrying certain genomic modifications (for instance, CDKN2A, aneuploidy) [65]. Hwang et al. included low CNA cells within the epithelial compartment that co-expressed ductal and follicular lineage markers, and these cell subpopulations were defined as transitional state cells (ADM and non-classical ductal cells). An inferred pseudotemporal trajectory from acinar to ADM to atypical ductal to malignant cells was inferred using the inferred pseudotemporal trajectory and found to parallel the monotonic increase in positive regulation of KRAS signaling, supporting ADM and atypical ductal cells as relevant intermediate states in pancreatic cancer tumorigenesis. Hwang et al. identified a set of low CNA nuclei within the epithelial compartment in the presence of co-expressed ductal and follicular lineage markers, which may reflect ADM. In addition, a distinct subpopulation of ductal cells express high levels of ductal (e.g., CFTR) and malignant (e.g., KRT5 and KRT19) markers without elevated CNA, which we termed atypical ductal cell expression, if also expressing HG PanIN signature genes (e.g., KRT17) is represented, then ADM converts to an atypical ductal state.

Another part argues that conversion from ADM is also required to undergo chemotaxis of receptor cells, which subsequently mutate before entering the oncogenic process. For example, partial conversion of adenoidal blast cells to HNF1B+ or POU2F3+ ADM populations leads to neoplastic transformation and formation of MUC5AC+ gastric-pit-like cells, including EEC, tufts cells, etc. Subsequent KRAS mutations activate cell differentiation to cancer epithelium [66,75]. DCLK1-positive cells represent a reservoir of pancreatic progenitor cells, which can maintain epithelial and in vivo follicular regeneration after tissue damage, and these cells are prone to develop into pancreatic cancer [110].

It is important to note in particular that although single cells allow us to gain insight into many transitional state cells and precancerous lesions of pancreas, we need to view these results with caution. Although many of the single-cell sequencing studies investigating ADM in recent years have claimed to use algorithms or manual single-cell removal of apparent doublets, coupled with spatial validation, there is still a significant false-positive rate. Because of similar ductal-acinar adhesion, cell in cell [112,113] can also present false ADM lesions, these effects cannot be fully removed by single-cell dissociation techniques and algorithms [114,115]. Therefore, based on the fact that pancreatic cancer single cells in particular can present false ADM lesions, the results of ADM analysis based on single cells of pancreatic cancer in particular still need to be clarified by subsequent experiments.

3 High risks precursor diseases: inflammation, precursor lesions and neoplasms

3.1 Chronic pancreatitis

Chronic pancreatitis is a disease caused by progressive inflammation and irreversible fibrosis caused by cumulative damage to pancreas over time. Excessive extracellular matrix deposition eventually leads to failure of internal and external hormone secretion. It usually presents with recurrent episodes of abdominal pain or pancreatitis and can progress to pancreatic cancer. A large subgroup of patients with neither pain (nearly 30%) nor a previous diagnosis of acute pancreatitis (about 50%) were also classified as having chronic pancreatitis. Chronic pancreatitis is usually associated with alcohol consumption, smoking or genetic risk factors, which notably increases the risk of pancreatic cancer. Sporadic chronic pancreatitis patients have a cumulative risk of 1.8% and 4% at 10 and 20 years, respectively, while those with hereditary pancreatitis have a 7.2% risk by age 70 years [116,117]. The annual incidence is 5 to 8 per 100 000 adults with a prevalence of 42 to 73 per 100 000 [116]. Common genetic mutations linked to chronic pancreatitis include CFTR, SPINK1, and CTRC, with over 90% developing early-onset pancreatic cancer [118]. Mouse models edited for genes like Spink1, Prss3b, and Cpa1 mirror the progression seen in humans with germline mutations [119121]. Hereditary pancreatitis, accounting for about 1% of all cases [116], is an autosomal dominant condition resulting from PRSS1 gene mutations. Despite its origin, chronic pancreatitis invariably increases pancreatic cancer risk. No consensus exists for cancer surveillance, but experts underline the importance of thorough monitoring, especially if symptoms like unexplained weight loss or new-onset diabetes are observed [122].

Chronic pancreatitis can cause increased susceptibility to pancreatic cancer by inducing ADM or by generating susceptible precursor cells. Preneoplastic pancreatic alterations have been previously identified in acute and chronic pancreatitis [123]. ADM rapidly shut down pancreatic enzyme expression, which may be able to limit the adverse effects of repetitive pancreatitis, allowing pancreatic tissue to recover rapidly from inflammation. However, persistent inflammatory stimulation of chronic pancreatitis results in extensive ADM persistance without recovery, increasing the risk of carcinogenesis. Any genetic event that promotes or stabilizes ADM, such as activating mutations in KRAS, may lead to the eventual progression to pancreatic cancer [110]. Tuft cells, a cancer precursor cell type, were commonly discovered in chronic pancreatitis and pancreatic cancer [65,66]. Also, reprogramming of DCLK1-positive cells may play an important role in inflammation-promoted tumorigenesis [110], which are present in chronic pancreatitis [124126].

3.2 Precursor lesions and neoplasms

The current WHO classification of tumors of the digestive system recognized five types of potential pancreatic cancer precursors: PanIN, mucinous cystic neoplasm (MCN), intraductal oncocytic papillary neoplasm (IOPN), intraductal tubulopapillary neoplasm (ITPN), and IPMN [59]. Fig.1 shows the common driving mutation genes of precancerous lesions, and the proportion of pancreatic cancer derived from each lesion. Most pancreatic precancerous lesions are usually macroscopic lesions > 0.5 cm, while PanIN is a microscopic lesion < 0.5 cm. In the past, the precancerous lesions were graded into three grades (low, moderate, and high). There was a general lack of consensus diagnostic markers to distinguish low-grade (LG) nonmalignancy from high-grade (HG) malignancy. Those previously classified as LG and “moderate grade” are now classified as LG, and those previously classified as HG remain the same. Many precancerous lesions are found incidentally after surgery. Despite the availability of laser capture microdissection techniques, the study of precancerous lesions in pancreatic cancer is still lacking. Nowadays, the availability of spatial omics allows obtaining microscopically diagnostic-dependent lesions for study using FFPE samples, which has significantly improved the understanding of pancreatic lesions (Tab.1).

3.2.1 IPMN

IPMN is defined as an apparently visible (usually > 5 mm), predominantly shaped or rarely flattened, noninvasive, mucin-producing epithelial tumor in the main ducts or branch ducts. The risk factors for the development of IPMN are unclear. The majority of IPMNs are located in the head of pancreas, and 40% of cases are multicentric. Median age of patients with IPMNs with invasive carcinoma is 3–5 years older than that of those without invasive carcinoma, indicating that progression to cancer takes time. IPMNs can be classified as “main duct type,” “branch duct type,” and “mixed duct type” according to their relationship with the main pancreatic duct. The main pancreatic duct type IPMN have higher risk of carcinogenesis than the other types. Mural nodules and/or irregular ductal contours may suggest HG neoplasms or invasive carcinoma.

Pathological subtypes of IPMN are divided into gastric type, intestinal type, and pancreatobiliary type. Gastric-type IPMNs are usually LG, intestinal type may be low or HG pancreatobiliary IPMN [127]. Intestinal-type IPMNs exhibit biological variances from gastric or pancreatobiliary types, encompassing disparities in the frequency of driver mutations (detailed in subsequent sections) and in the predominant and branching duct locations. Somatic alterations in hotspots of the KRAS oncogene represent some of the initial genetic shifts in IPMNs, whereas mutations in CDKN2A, TP53, and SMAD4 arise at more advanced stages (refer to Fig.1). Furthermore, genomic analyses of IPMNs have uncovered additional driver genes uniquely characterized by a high mutation frequency confined to IPMNs. For example, mutations in GNAS are common in IPMN, which are thought to be alternative triggers for KRAS tumor formation. The prevalence of GNAS mutation varies by pathological subtypes, and thus the prevalence of GNAS mutation reported in IPMN cohorts varies, ranging from 40% to 80%. Most intestinal IPMNs are predominantly ductal lesions with a high frequency of GNAS mutations. Furthermore, RNF43, a tumor suppressor gene, frequently exhibits inactivating mutations in IPMNs, believed to surface following initial alterations in KRAS and GNAS. While RNF43 mutations are sparingly seen in pancreatic cancer, their significant occurrence is specific to IPMNs. Another distinctive prospective driver in IPMN development, KLF4, has been pinpointed in association with low-grade (LG) IPMNs. In-depth multi-regional and single-cell sequencing investigations into IPMNs have uncovered considerable genetic diversity in driver mutations. Interestingly, LG IPMNs often harbor several distinct KRAS or GNAS mutations present in separate cells, delineating various clones devoid of other somatic mutations. This implies a potential polyclonal origin for many IPMNs. PanIN and gastric IPMN are distinguished based on morphology and size and show the same immunohistochemical profile, with diffuse positivity for MUC5AC, without MUC1 and MUC2 expression. Intestinal IPMN are distinctly different lesions at both morphological and immunohistochemical levels and are characterized by positive MUC2 and MUC5AC [128]. A previous study found that in approximately 20% of cases, concurrent IPMN and adenocarcinoma were independent of each other, and IPMN proved to be a mere parallel event in a subset of pancreatic cancers [129,130]. The study found that IPMN and adenocarcinoma were independent of each other in about 20% of cases, and IPMN proved to be a simple parallel event in a proportion of pancreatic cancers. The unique epigenetic control and expression patterns of MUCL3, coupled with frequent copy number variations (primarily deletions) in gastric IPMNs, could signal a heightened propensity for these lesions to progress. Intestinal IPMNs, characterized by a distinct genetic composition and elevated genomic instability, manifest higher proliferation rates even in lower-grade lesions, indicating a potentially greater risk of progression compared to PanINs and gastric IPMNs. Unlike PanINs and gastric IPMNs, intestinal IPMNs exhibit an increased presence of genes related to mucin secretion and distinctly divergent epigenetic landscapes informed by DNA methylation patterns, which correlate them with various mature cell types in the ductal array. Histopathological subtyping employs markers like MUC1, MUC2, MUC5AC, and CDX2. The spatial transcriptome of IPMN is only in its infancy, and some unpublished preprints observed a high degree of heterogeneity in expression profiles and mucins at different pathological differentiation directions and degrees of differentiation in gastric, intestinal, and pancreaticobiliary types, suggesting that IPMN still has the potential to be subdivided [72,73].

3.2.2 ITPN

In more than 75% of ITPN cases, adenocarcinoma is also simultaneously found [131]. However, ITPN accounts for only 3% of pancreatic precancerous lesions and only 0.9% of all exocrine malignancies (Fig.1) [132]. The most common location of ITPN is head of pancreas, which more often involved the main pancreatic duct/Wirsung duct (72.5%), or both main and branch pancreatic ducts (10%). Only 17.5% ITPN were observed along the branch pancreatic ducts [133]. Although associated invasive carcinoma is common, the prognosis of ITPN is much better than pancreatic cancer, with a 5-year survival rate of 71% in patients with ITPN-associated invasive carcinoma. Histologically, largely considered HG ITPN has a unique form of mucin expression, positive for MUC1 (> 90%) and MUC6 (70%), while lower levels of expression for MUC2 (8.6%), MCL amplifications (31.8%), FGFR2 fusions (18.2%), PI3KCA mutations (13.6%) are more common in ITPN (P < 0.001) [133]. The mechanism of progression of ITPN to pancreatic cancer is currently unclear, and Fukunaga et al. induced deletion of Arid1a and Pten in pancreatic ductal cells through activation of the YAP/TAZ pathway, leading to ITPN and ITPN-associated pancreatic malignancies [52]. ITPN most likely originates in the pancreatic ducts, unlike pancreatic cancer which originates in the classical pancreatic acinar cells. It also possesses rather different mutant forms [134]. Common pancreatic cancer driver genes (e.g., KRAS mutations (10.4%), TP53 mutations (4.7%), CDKN2A mutations (27.2%), and the gene SMAD4 mutations, GNAS mutations, RNF43 mutations) happened to have less mutations rates in ITPN [133]. In contrast, the relative lack of pancreatic cancer alterations in this case series highlights the molecular differences with conventional pancreatic cancer, but it should be acknowledged that KRAS alterations are still present in a non-negligible set of cases (still observed in 25% of patients with classical KRAS mutations) [131]. Approximately 30% of ITPNs have activating mutations in genes involved in the PI3K/Akt pathway (PIK3CA, PIK3CB, INPP4A, PTEN) and chromatin-remodeling factors (MLL1, MLL2, MLL3, BAP1, PBRM1, EED, ATRX, ARID2, ASXL1), suggesting that ITPN harbors distinct genetic alterations [135]. Some chromatin-remodeling factors were present only in the infiltrative component. As for chromosomal alterations, 1q increases (75%) and 1p, 6q or 18q deletions (about 50%) are the most common. As for structural variants, common fusions involve the recently identified RET and FGFR2. All ITPN and concurrent adenocarcinomas have most of the same genomic alterations [135].

3.2.3 MCN

MCNs of the pancreas are recognized for their mucin-producing capabilities and potential for malignant transformation. Their classification, mirroring PanINs and IPMNs, relies on the architectural and cytological atypia, segmenting them into low-grade (LG) or high-grade (HG). While LG MCNs predominantly feature gastric foveolar differentiation, HG MCNs exhibit pancreatobiliary traits. Notably, MCNs do not manifest the intestinal differentiation common in IPMNs. There is the absence of connections to the pancreatic duct system and the presence of a unique ovarian-type stroma beneath their neoplastic epithelium. This stroma is a diagnostic hallmark of MCNs. Interestingly, genetic investigations suggest an origin of MCNs from halted primordial germ cells, rather than standard pancreatic cells, elucidating their distinct cellular beginnings. This theory aligns with the clinical observations that MCNs predominantly affect females. From a genetic standpoint, early-stage MCNs frequently harbor mutations in the KRAS oncogene. Yet, as they progress, other genetic anomalies become evident. For instance, RNF43 mutations are prevalent in MCNs, reminiscent of IPMNs. However, GNAS mutations, a characteristic of IPMNs, remain absent in MCNs, highlighting their genetic disparity [136,137].

3.2.4 IOPN

IOPNs consist of complex dendrites lined with multiple layers of cuboidal cells with granular eosinophilic granular stroma, round nuclei and prominent nucleoli. IOPNs were once classified as a subtype of IPMN, and they are now considered to be a distinct tumor. They are almost universally considered to have HG heterogeneous proliferation, and associated invasive carcinomas are common. But patients with IOPN are usually small and have a longer survival than pancreatic cancer. IOPNs lack somatic mutations in typical IPMN driver genes, but have been shown to have gene fusions involving PRKACA or PRKACB (Fig.1) [138,139]. IOPNs are > 1 cm cystic nodular lesion with oncocytic features and ductal differentiation and are associated with pancreatic cancer in 60% of the cases, and IOPNs are frequently HG [127]. Detailed general descriptions of the tumors are available, most of which are described as multicompartmental or uni-compartmental cysts, some of which contain papillary projections or solid nodules. About 50% tumor is clearly associated with the main pancreatic duct. Microscopically, the tumor appeared as multifoveal or unifoveal cysts, with multifoveal and heterogeneous gross appearance. The tumor is arranged by multiple layers of tumor cells with abundant granular eosinophilic cytoplasm and large, fairly homogeneous nuclei containing a single distinct nucleolus. The 10-year overall survival of the whole cohort was 94%, and there was no difference between the invasive and noninvasive IOPN cohorts (P = 0.38) [140].

3.2.5 PanIN

Unlike other precancerous lesions of pancreatic cancer IPMN and MCN, both of which are macroscopic lesions, PanIN is defined as a microscopic, flat or shaped, noninvasive epithelial tumor that is < 5 mm in diameter on standard histological sections stained with hematoxylin and eosin and is characterized by varying amounts of mucin as well as cytological and structural degree of heterogeneity. It is considered to be the most common precancerous lesion of pancreatic cancer. However, the risk factors for PanIN are presumed to be similar to those for invasive pancreatic cancer. Thus, they would include old age, smoking, obesity, long-term diabetes, and chronic pancreatitis. Recent studies in mice have shown that pancreatic acinar cell carcinoma origin forms PanIN, whereas adenocarcinoma of ductal origin is not associated with the formation of PanIN. It is also possible to distinguish pancreatic cancer of ductal and follicular origin with high AGR2 [141].

Telomere shortening and activation site mutations in the KRAS oncogene appear to be among the earliest genetic alterations in PanIN lesions, as these genetic changes are present in most PanIN with LG heterogeneous hyperplasia. Inactivating mutations in the CDKN2A gene begin to appear in PanIN with low- to moderate-grade heterogeneous hyperplasia, and SMAD4 and TP53 mutations are common in HG PanIN, as they are largely only seen in PanIN with HG heterozygosis. The earliest alterations in pancreatic tumorigenesis are KRAS oncogenic hotspot mutations and telomere shortening, and a high prevalence occurs even in early lesions, with > 90% of LG PanINs having KRAS hotspot mutations and short telomeres. In contrast, the prevalence of alterations in key tumor suppressor genes (e.g., CDKN2A and TP53) in pancreatic tumorigenesis increases with increasing grade of heterozygosity, and the uniqueness of SMAD4 mutations in the landscape of pancreatic cancer is underscored by their rarity in PanINs, distinguishing them as a late-emerging genetic driver in the disease’s progression. However, the absence of such mutations in a substantial percentage of pancreatic cases highlights the complexity and variability in the genetic underpinnings of this malignancy. PanINs may be intraductal extensions of invasive tumors and can be shown to be remote from the primary tumor in large numbers and discontinuously. HG PanINs are phylogenetically associated with infiltrative cancers and contain as many base substitutions as possible, but with fewer copy number alterations [142]. This can be explained by the colonization of PanINs through so-called ductal carcinogenesis, a phenomenon now reported clinically using histology [127,143]. The prevalence of chromosomal alterations and chromosome splitting increases with increasing PanIN grade, suggesting that aneuploidy may be a useful co-biomarker for driving mutations. In recent years, with the use of single-cell and spatial histology, the characteristics of PanIN are expected to be further revealed, with PanIN exhibiting increased expression of extracellular matrix-related genes (DCN, SPARC, and SPON1), a diversity of collagens, genes involved in ADM reprogramming (KLF4 and MMP7) and other markers of early-stage malignancy (CXCL12, TIMP3, ITGA1, and MUC5AC) [64]. The prevalence of LG precursor lesions is high, and in a recent Japanese autopsy series, LG PanINs were found in more than 75% of the population, and HG PanINs were found in approximately 5% of patients [144]. There was no difference in the prevalence of precursor lesions in patients with or without germline susceptibility [145]. As precancerous lesions can be completely removed surgically, PanINs represent promising potential target lesions for resection. However, there are challenges for lacking early detection methods.

4 Biomarkers

Pancreatic cancer is a progressive disease influenced by genetic mutations and epigenetic changes. From its initial mutation to clinical presentation, pancreatic cancer can evolve over a span of 15–20 years [146]. However, the absence of accurate diagnostic techniques underscores the need to identify appropriate biomarkers for pancreatic cancer prevention and treatment. In recent years, advancements in artificial intelligence algorithms and various detection methods have significantly improved the discovery and utilization of biomarkers for pancreatic cancer. Recent advancements in artificial intelligence algorithms and diverse detection methodologies have greatly enhanced the identification and application of biomarkers in pancreatic cancer.

An ideal biomarker should offer consistent and effective diagnostic capabilities, be non-invasive, and monitor disease progression in real time [147]. For pancreatic cancer, CA19-9 stands as the sole United States Food and Drug Administration (FDA)-approved biomarker. Yet, it lacks both sensitivity and specificity, notably among patients with pancreatitis and those devoid of Lewis antigen expression [148]. Pathological criteria currently serve as the gold standard for diagnosing pancreatic cancer. Given the pancreas’s distinct anatomical position and the challenges of replicating invasive procedures, choosing the right biomarkers has become a pivotal focus in contemporary pancreatic cancer research. Presently, sources such as blood, urine, pancreatic fluid, and gut microbiota offer potential biomarkers for pancreatic cancer. These biomarkers include various biological sample components like nucleic acids (for instance, circulating tumor DNA (ctDNA), cell-free circulating DNA (cfDNA), microRNAs (miRNA)) and their modifications (like DNA methylation), as well as proteins, peptides, cells (such as CTCs), and extracellular vesicles (like exosomes) [147,149]. Concurrently, recent research indicates that individual biomarkers’ diagnostic capabilities remain insufficient. Pairing them with CA19-9 can boost their efficacy, which is a practice frequently adopted in current studies (Tab.3).

4.1 Blood biomarkers

Blood receives raw materials, secretions, and waste products emitted by tumor cells. Thus, changes in blood composition are linked to the pathological processes of tumor cells. Blood is the most extensively researched source for biomarkers, encompassing types such as nucleic acids, proteins, lipids, and polysaccharides.

In recent years, there has been significant research on blood nucleic acids as potential biomarkers for pancreatic cancer. DNA fragments known as cfDNA are those present in blood’s non-cellular components. They can result from the apoptosis and necrosis of tumor cells or healthy cells, as well as from the direct secretion of tumor cells or other microenvironmental cells, such as immune and inflammatory cells. The variable component of cfDNA produced by apoptotic or necrotic tumor cells is sometimes referred to as ctDNA, and it is recognized by certain cancer-related mutations [150,151]. Since ctDNA is directly derived from tumor cells, it has a high specificity and is significantly linked to tumor metastasis [150,151]. As a result, it is a valuable diagnostic tool that can predict a patient’s prognosis. Although tumors have more cfDNA than healthy individuals, cfDNA detection is still tricky. To identify pancreatic cancer, the researcher applied the detection of cfDNA as a biomarker and paired it with CA19-9 and THBS2 (thrombospondin-2), which can achieve an impressive area under curve (AUC) of 0.94 [150]. Since KRAS mutation is prevalent in more than 90% of pancreatic cancer cases, ctDNA carrying the KRAS mutation has been extensively studied as a biomarker. Research has demonstrated that detecting KRAS mutation ctDNA is highly effective in predicting chemotherapy response and monitoring disease recurrence [152]. Recent investigations have indicated that mRNA [153,154], non-coding RNA (ncRNA) [146,155157], tRNA-derived small RNA (tsRNA) [158,159], and other RNAs have high potential as biomarkers. Some of these RNA molecules are found in extracellular vesicles, which enhances their abundance and integrity as diagnostic markers. Moreover, the detection of nucleic acid alterations has emerged as a potent method in pancreatic cancer diagnosis. For instance, Majumder et al. developed a methylation DNA assay in combination with CA19-9, achieving an impressive AUC value of 0.97 [160].

With advancements in mass spectrometry analysis and enhanced detection techniques, our ability to detect various metabolites, including proteins, lipids, and oligosaccharides, has seen significant improvement recently. And research on the emergence of pancreatic cancer biomarkers based on this has also significantly increased.

The pancreatic cancer diagnosis model created by Kim et al. [161] and Mahajan et al. [162] utilized metabolic proteins and had an excellent AUC value. Asprosin was also employed by Nam et al. to efficiently and accurately diagnose pancreatic cancer [163]. The potential of lipids as a biological diagnostic tool for pancreatic cancer is very broad because the lipid changes may result from the tumor and tumor microenvironment cells and could also reflect the organism’s immune response. Using the lipid model, Wolrab et al. can correctly identify pancreatic cancer [164]. CA19-9, an oligosaccharide secreted by tumor cells, has been approved by the FDA as a biomarker for pancreatic cancer. Yet, elevated levels of this substance are not universal among all patients. Considering this limitation, researchers have investigated an alternative oligosaccharide known as sialylated tumor-related antigen (sTRA) for its potential diagnostic replacement for CA19-9. The study findings suggest that sTRA exhibits comparable diagnostic potential to CA19-9 in clinical patients [165].

4.2 Urine biomarkers

Although urine biomarkers have received less attention as a diagnostic method for pancreatic cancer, they hold significant promise. The kidneys’ ultrafiltration function makes urine an advantageous source for biomarker analysis, offering benefits such as easy sample collection, reproducible procedures, higher concentration, and reduced variability. Notably, Debernardi et al. has developed a diagnostic model for pancreatic cancer utilizing three proteins that exhibit stable expression in urine and bind to CA19-9, yielding an impressive AUC of 0.992 [166].

4.3 Pancreatic fluid biomarkers

Pancreatic fluid holds great potential as a source of biomarkers for pancreatic cancer due to its proximity to the ductal cells that produce PDAC. As the pancreas directly secretes pancreatic fluid, it will likely contact tumor cells directly, resulting in a higher specificity than other sample sources. With advancements in duodenoscopy and endoscopic ultrasound (EUS), obtaining pancreatic fluid has become safer. Nesteruk et al. discovered that pancreatic fluid exhibited higher sensitivity than serum for pancreatic cancer diagnosis. In addition, they found that miRNA present in extracellular vesicles of pancreatic fluid combined with CA19-9 was effective in diagnosing pancreatic cancer with a diagnostic AUC value of 0.91 [167].

4.4 Gut microbiota biomarkers

The connection between gut microbiota and the pancreas is evident, with notable alterations in the gut microbiota of pancreatic cancer patients compared to healthy individuals. These changes in gut microbiota are intricately involved in the progression of pancreatic cancer, highlighting its potential as a biomarker for this disease. Kartal et al. developed a predictive model utilizing 27 species of gut microbiota combined with CA19-9, which achieved an impressive AUC of up to 0.94 for pancreatic cancer detection [168]. Furthermore, Nagata et al. employed machine learning algorithms to establish a diagnostic model for pancreatic cancer based on gut microbiota, demonstrating significant diagnostic efficacy. This model was further validated in Germany, Spain, and Japan [155].

5 Chemoresistance

Given the challenges in diagnosing pancreatic cancer early on, a majority of patients are identified with advanced tumors or metastases, leaving only about 20% with surgical options [3]. Most patients require chemotherapy, among which GEM-based chemotherapy is the first-line chemotherapy regimen for pancreatic cancer [169,170]. However, chemoresistance limits the clinical outcomes of chemotherapy (Fig.2).

5.1 TME

The TME plays a pivotal role in the formation and progression of tumors. Specifically, the TME of pancreatic cancer is marked by a dense intercellular matrix and a notable accumulation of extracellular matrix [171]. Pancreatic cancer cells are encapsulated within a thick fibrotic matrix [172]. Excessive fibrosis around the tumor cells and a large majority of interstitial components increases the interstitial fluid pressure (IFP), reaching more than 10 times higher than in healthy pancreas. The formation of a high-pressure barrier around the tumor cells prevents chemotherapy treatments from reaching the tumor cells because it reduces blood flow to the tumor cells, results in blood vessel collapse, and inhibits the penetration of chemotherapy drugs [173]. Additionally, this situation exposes tumor cells to ischemic, hypoxic, and acidic conditions, triggering their metabolic reprogramming and elevating their resistance to chemotherapy [171].

Beyond causing physical alterations due to elevated interstitial pressure and its ensuing effects, interstitial elements in the tumor microenvironment also promote the chemotherapy resistance of tumor cells, including fibroblasts, pancreatic astrocytes, extracellular matrix, and immune cells [174].

By secreting exosomes and cytokines, interstitial cells help tumor cells develop resistance to chemotherapy [175]. By secreting stromal platelet-derived growth factor receptor (PDGFR), stromal fibroblasts cause interstitial contracture and exacerbate interstitial hypertension [176]. Hu et al. discovered that CAFs elevate LIF secretion, thereby activating the STAT3 signaling pathway in pancreatic cancer cells [177]. This increase heightens the expression of proteins tied to GEM resistance like ABCC2, CDA, and SOX2, leading to the rise of GEM-resistant pancreatic cancer cells [177]. Additionally, CAFs can release TGF-1, IL-8, CXCL12, and other substances that activate the SMAD2/3, NF-κB, and other chemotherapy resistance-related pathways [178180]. Besides, M2 macrophages can generate extracellular vesicles, which can be used to deliver miR-222-3p to tumor cells and activate the PI3K/AKT/mTOR pathway, which increases chemoresistance [181]. Tumor-associated macrophages (TAM) can induce GEM resistance by secreting pyrimidine and competitively inhibiting GEM [182]. Laminin (LN), a protein of the extracellular matrix, induces FAK phosphorylation in pancreatic cancer cells, resulting in Akt phosphorylation and increased expression of apoptosis-related proteins (increasing survivin and pBad (pS136) levels), thereby inducing GEM resistance in pancreatic cancer [183].

Moreover, upon exposure to chemotherapy, these resistant cells further drive the emergence of chemotherapy-resistant counterparts through the secretion of exosomes and cytokines. Richards et al. discovered that GEM treatment could increase the expression of Snail and Snail target microRNA-146a in CAF and transfer these proteins to tumor cells through exosomes to promote GEM-resistance in tumor cells [184]. Richards et al.’s study found that CAFs exposed to GEM release exosomes containing miRNAs (miR-21, miR-181a, miR-221, miR-222, and miR-92a) that act on PTEN in tumor cells, which can lead to chemoresistance [185]. This interplay of chemoresistance is also evident between tumor cells. Patel et al. observed that upon GEM treatment, pancreatic cancer cells amplify exosome secretion. This results in the transfer of miR-155 to drug-sensitive pancreatic cancer cells, subsequently suppressing DCK expression and fostering chemoresistance [186].

5.2 Formation of chemoresistance mechanism in pancreatic cancer

Pancreatic cancer may experience mechanism modifications to formate chemoresistance in addition to TEM alterations in pancreatic cancer [187]. Recent studies have revealed that chemoresistance can arise from cancer stem cell (CSC), EMT, and epigenetics. According to Cioffi et al.’s research, the percentage of CSC is inversely correlated with the tumor’s chemoresistance, and some treatment strategies specifically targeting CSC are more effective in preventing the development of chemoresistance [188,189]. Xiaofeng Zheng suppressed EMT by creating transgenic mice with EMT-induced transcription factor (Snail or Twist) knockouts and discovered that balance nucleoside transporter (ENT1) and concentrated nucleoside transporter (Cnt3) were dramatically elevated after EMT inhibition, and it can significantly increase the sensitivity of GEM [190]. m6A methylation stands out as the predominant epigenetic modification. GEM chemoresistance can be prevented by ALKBH5, an m6A demethylase, via transactivating WIF-1 and subsequently inhibiting Wnt signaling [191]. By facilitating the upregulation of DDIT4-AS1 by activating the mTOR pathway, it can also preserve the stemness of pancreatic cancer and decrease chemosensitivity.

Analyzing the differentially expressed genes (DEGs) across pancreatic cancer cell lines with distinct drug resistance profiles revealed that these DEGs predominantly modulate GEM metabolism transporters, cell cycle regulation, and signaling pathways associated with proliferation or apoptosis [192]. Furthermore, a spectrum of ncRNAs, encompassing long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), and miRNAs, have emerged as regulators of chemoresistance in pancreatic cancer. As highlighted by Gu et al., the elevated expression of ABC transporter proteins, steered by the hsa-miR-3178/RhoB/PI3K/Akt axis, culminates in GEM resistance in pancreatic cancer cells [193]. According to Tsai-Fan Chou, lncRNA PVT1 enhances autophagy activity and Wnt/β-catenin pathway signaling to promote GEM resistance in pancreatic cancer [194]. lncRNA GSTM3TV2 competitively regulates let-7, upregulating LAT2 and OLR1, thereby increasing GEM resistance in pancreatic cancer [195]. The Circ-MTHFD1L/miR-615-3p/RPN6 signaling axis maintains GEM resistance in pancreatic cancer [196]. Upregulation of vASH2 leads to JUN induction, which activates RRM2 transcription through direct binding to the RRM2 promoter, resulting in GEM resistance [197]. Furthermore, researchers have found that GEM increases the expression of cyclin D1 and TAK1 in pancreatic cancer, promoting cell proliferation and inhibiting drug-induced apoptosis, consequently leading to chemoresistance [198,199]. Activation of the EGFR self-activation (phosphorylation), mTOR signaling pathway, NF-κB signaling pathway, and STAT3 signaling pathway in pancreatic cancer also plays a crucial role in promoting the development of chemoresistance [200,201].

6 Immunotherapy resistance

Immunotherapy is a new direction of tumor therapy. Currently, immunotherapy encompasses adoptive cell therapy (such as chimeric antigen receptor T cells (CAR-T)) [202,203], oncolytic vaccines, and immune checkpoint blocking (ICB) [204]. Immunotherapy has got positive survival outcomes in patients with advanced solid malignancies, such as melanoma and lung cancer. However, directly transplanting this therapeutic approach is not effective for pancreatic cancer, a challenge attributed to the immunotherapy resistance of this cancer type (Fig.3).

Pancreatic cancer is characterized by its low immunogenicity and suboptimal antigen presentation [205]. The density of its tumor-associated antigens (TAAs) is markedly diminished compared to malignancies like melanoma, and its tumor mutation burden (TMB) also lags, posing significant challenges to igniting anti-tumor immune responses [206,207]. Additionally, the lack of mature DC cells in pancreatic cancer can lead to poor antigen presentation, which inhibits T cell activation [208].

Concurrently, there is a notable redistribution of immune cells within the TME of pancreatic cancer [209]. T lymphocytes play an essential role in anti-tumor immunity, and T cells in TME include CD8+ cytotoxic T cells (CTL), also known as effector T cells [210]. CD4+ T cells include helper T cells (Th) Th1, Th2, Th17, Tregs. CTL and Th1 CD4+ T cells are conducive to anti-tumor immunity. However, in TMEs of pancreatic cancer, the distribution of CTL is infrequent, and this scarcity becomes increasingly pronounced as one approaches the tumor [206]. Furthermore, the TME experiences an influx of numerous immunosuppressive cells, encompassing entities like TAMs, myeloid-derived suppressor cells (MDSCs), and Tregs, etc. [211], which can inhibit the body’s anti-tumor immune response by inhibiting the killing function of natural killer (NK) cells, DCs, effector T cells, and other cells. At the same time, these immune cells will appear to crosstalk [212]. The net result further inhibits the distribution and infiltration of immunosuppressive effector cells and increases the proportion of immunosuppressive cells [207].

TME stromal cells and pancreatic cancer cells can release cytokines that alter immune system components. To be specific, KRAS-mutated tumor cells have the capacity to upregulate granulocyte-macrophage colony-stimulating factor (GM-CSF) and MDSC chemotaxis [213]. CCL2 secreted by pancreatic cancer promotes macrophage infiltration [214]. CXCL1 secreted by tumor cells can cause an increase in bone marrow cells and a decrease in cytotoxic CD8+ T cell infiltration [215]. In addition, CAFs can also mediate the upregulation of immune checkpoints in T cells to inhibit adaptive immunity and can secrete macrophage colony-stimulating factor (M-CSF) to promote M2 polarization of macrophages [216] and promote the recruitment of MDSC in a CCR2-dependent manner [217].

7 Treatment of pancreatic cancer

The outlook for pancreatic cancer remains deeply concerning. Although the basic research and treatment methods of pancreatic cancer have made breakthroughs in recent years, the 5-year survival rate is only 10%. At present, surgical resection combined with systemic chemotherapy is the only way for pancreatic cancer patients to survive for a long time. And the main therapeutic methods for pancreatic cancer patients are surgical therapy, chemotherapy, targeted therapy, immunotherapy, etc. [218]. At the same time, the therapeutic concept is developing in the direction of multi-discipline and individual.

Owing to the pancreas’s intricate anatomical position, pancreatic surgery presents formidable challenges. Undertaking a surgical resection for pancreatic cancer demands unparalleled surgical acumen and extensive perioperative care expertise [219]. Perioperative mortality has been lowered to 3%–4% owing to ongoing advancements in pancreatic surgery methods and techniques [220]. As surgical protocols for pancreatic cancer enhance safety, a broader cohort of patients now becomes eligible for surgical intervention compared to the past. Recent research has shown that surgical treatment, particularly the excision of R0, can improve survival outcomes in patients with pancreatic cancer who have locally advanced stage [221], borderline resectable [222], and distant oligo metastases (such as liver/distant lymph node metastasis) [223,224]. Due to pancreatic surgery’s high difficulty and complications, many medical centers will prudently select surgical patients, especially some older adults. However, the latest research shows that aging does not affect the long-term survival of pancreatic cancer patients undergoing surgical treatment [225]. At the same time, the emergence of neoadjuvant therapy can transform some patients from unresectable to having the chance of surgery, realizing the downtime, and improving the proportion of R0 excision [221,222].

The impact of surgical interventions and associated complications on the duration and efficacy of chemotherapy has remained a subject of ongoing debate. The recent ESPAC-3 study indicated that, as long as patients can finish the entire chemotherapy plan, their prognosis will not be impacted even if postoperative chemotherapy is started 12 weeks after surgery instead of the traditional 6 weeks after surgery [226]. Advancements in minimally invasive techniques, encompassing laparoscopic and robotic procedures, promise reduced patient discomfort, shorter hospital stays, and expedited recovery, all while maintaining surgical safety [227229].

Over the past several decades, GEM has emerged as the cornerstone chemotherapy regimen for pancreatic cancer patients. With the deepening of research in recent years, the current first-line chemotherapy regimen is FOLFIRINOX and GEM plus albumin combined with paclitaxel [230]. However, there is no doubt that the side effects of chemotherapy and chemoresistance limit the prognosis of patients with pancreatic cancer. At present, some chemotherapy-based combination therapy strategies have been proposed with encouraging results, but it will take a significant amount of time to clinically verify these schemes (Tab.4).

The intricate pathological landscape of pancreatic cancer is predominantly governed by gene mutations, which hold pivotal roles in the disease’s onset and progression. Currently, KRAS, TP53, CDKN2A, and SMAD4 have been identified as the four primary driving genes involved in the whole process of disease development [231]. Additionally, Sian Jones reported 12 core signaling pathways enriched in mutated genes in pancreatic cancer and proposed that regulating these signaling pathways is also a direction for treatment [232]. All these also provide a theoretical basis for the targeted therapy of pancreatic cancer (Tab.4). However, the progress of targeted therapy is limited in pancreatic cancer, and the vast majority of targeted therapy trials have failed, which is closely related to the complexity and compensation of mechanism regulation [233].

Immunotherapy is a new direction of current treatment, which has improved the survival of patients with advanced solid tumors, including melanoma and lung cancer [234236]. However, the outcomes of pancreatic cancer have been disheartening [218]. Current immunotherapy in pancreatic cancer includes immune checkpoint suppression therapy, pancreatic cancer immune vaccine, and CAR-T infusion, but these outcomes are poor [237,238].

Treatment with immune checkpoint inhibitors, including PD-L1 and CTLA-4, has failed but has shown promising results in a subset of patients with microsatellite instability, indicating a direction for immunotherapy in pancreatic cancer. Furthermore, based on the fact that previous immunotherapy has little clinical activity, there are also several multimodal immunotherapies that are being tried, such as chemotherapy/radiotherapy combined immunotherapy, and some good clinical results have been obtained [206,239].

8 Conclusions

As of now, the advancements in chemotherapy and targeted treatments have modestly elevated the 5-year survival rate for pancreatic cancer patients, rising from a mere 2% a decade ago to 12% in 2022. And our deepened comprehension of pancreatic cancer’s subtype biology paves the way for a more nuanced and targeted therapeutic approach (Fig.4). New clinical trial designs, including drug introductions, new adjunctive tests for investigational drugs, and platform studies that allow for rapid test combinations, are facilitating progress. Pancreatic cancer encompasses a diverse array of malignant epithelial neoplasms, characterized by intricate histological configurations and a varied genetic and molecular landscape. This malignancy arises from multiple precursor lesions, notably PanIN, IPMN, IOPN, ITPN, and MCN. The latest molecular categorization of pancreatic cancer, informed by comprehensive genomic, transcriptomic, proteomic, and epigenetic analyses, offers critical insights into its molecular diversity and the inherently aggressive nature of the disease. Investigations at the single-cell level have shown significant promise, surmounting earlier challenges arising from the amalgamation of stromal and tumor cells. However, there exist several pressing questions that still await comprehensive answers. While single-cell analyses have yet to establish a molecular classification that deeply informs clinical protocols, they are also hampered by the need for fresh tissue samples, affecting the quality and collaborative scope of research due to subpar cell dissociation and RNA integrity. This issue contributes to an insufficient capture of stromal cells in pancreatic cancer samples, skewing cell type representation. Though neoadjuvant therapies show promise, a significant knowledge gap persists as current genomic studies typically concentrate on pre-treatment conditions. Investigating the genomic consequences of treatment, particularly through comparative pre- and post-therapy mRNA studies, is crucial. The practical impact of molecular findings hinges on their clinical applicability, necessitating a focus on cell-specific biology, intercellular interactions, and tumor behavior over time to craft targeted treatment plans. Despite the nascency of precision oncology in pancreatic cancer, the anticipation is building for trials utilizing specific molecular categorizations and markers, signaling a shift away from uniform treatment methods.

References

[1]

Rahib L, Wehner MR, Matrisian LM, Nead KT. Estimated projection of US cancer incidence and death to 2040. JAMA Netw Open 2021; 4(4): e214708

[2]

Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin 2021; 71(1): 7–33

[3]

Mizrahi JD, Surana R, Valle JW, Shroff RT. Pancreatic cancer. Lancet 2020; 395(10242): 2008–2020

[4]

Park W, Chawla A, O’Reilly EM. Pancreatic cancer: a review. JAMA 2021; 326(9): 851–862

[5]

Huang J, Lok V, Ngai CH, Zhang L, Yuan J, Lao XQ, Ng K, Chong C, Zheng ZJ, Wong MCS. Worldwide burden of, risk factors for, and trends in pancreatic cancer. Gastroenterology 2021; 160(3): 744–754

[6]

Maisonneuve P, Lowenfels AB. Risk factors for pancreatic cancer: a summary review of meta-analytical studies. Int J Epidemiol 2015; 44(1): 186–198

[7]

Lynch SM, Vrieling A, Lubin JH, Kraft P, Mendelsohn JB, Hartge P, Canzian F, Steplowski E, Arslan AA, Gross M, Helzlsouer K, Jacobs EJ, LaCroix A, Petersen G, Zheng W, Albanes D, Amundadottir L, Bingham SA, Boffetta P, Boutron-Ruault MC, Chanock SJ, Clipp S, Hoover RN, Jacobs K, Johnson KC, Kooperberg C, Luo J, Messina C, Palli D, Patel AV, Riboli E, Shu XO, Rodriguez Suarez L, Thomas G, Tjønneland A, Tobias GS, Tong E, Trichopoulos D, Virtamo J, Ye W, Yu K, Zeleniuch-Jacquette A, Bueno-de-Mesquita HB, Stolzenberg-Solomon RZ. Cigarette smoking and pancreatic cancer: a pooled analysis from the pancreatic cancer cohort consortium. Am J Epidemiol 2009; 170(4): 403–413

[8]

Pang Y, Kartsonaki C, Guo Y, Bragg F, Yang L, Bian Z, Chen Y, Iona A, Millwood IY, Lv J, Yu C, Chen J, Li L, Holmes MV, Chen Z. Diabetes, plasma glucose and incidence of pancreatic cancer: a prospective study of 0.5 million Chinese adults and a meta-analysis of 22 cohort studies. Int J Cancer 2017; 140(8): 1781–1788

[9]

Canto MI, Almario JA, Schulick RD, Yeo CJ, Klein A, Blackford A, Shin EJ, Sanyal A, Yenokyan G, Lennon AM, Kamel IR, Fishman EK, Wolfgang C, Weiss M, Hruban RH, Goggins M. Risk of neoplastic progression in individuals at high risk for pancreatic cancer undergoing long-term surveillance. Gastroenterology 2018; 155(3): 740–751.e2

[10]

Corral JE, Mareth KF, Riegert-Johnson DL, Das A, Wallace MB. Diagnostic yield from screening asymptomatic individuals at high risk for pancreatic cancer: a meta-analysis of cohort studies. Clin Gastroenterol Hepatol 2019; 17(1): 41–53

[11]

Yuan C, Babic A, Khalaf N, Nowak JA, Brais LK, Rubinson DA, Ng K, Aguirre AJ, Pandharipande PV, Fuchs CS, Giovannucci EL, Stampfer MJ, Rosenthal MH, Sander C, Kraft P, Wolpin BM. Diabetes, weight change, and pancreatic cancer risk. JAMA Oncol 2020; 6(10): e202948

[12]

Chari ST, Leibson CL, Rabe KG, Ransom J, de Andrade M, Petersen GM. Probability of pancreatic cancer following diabetes: a population-based study. Gastroenterology 2005; 129(2): 504–511

[13]

Gupta S, Vittinghoff E, Bertenthal D, Corley D, Shen H, Walter LC, McQuaid K. New-onset diabetes and pancreatic cancer. Clin Gastroenterol Hepatol 2006; 4(11): 1366–1372

[14]

Munigala S, Singh A, Gelrud A, Agarwal B. Predictors for pancreatic cancer diagnosis following new-onset diabetes mellitus. Clin Transl Gastroenterol 2015; 6(10): e118

[15]

Duell EJ, Lucenteforte E, Olson SH, Bracci PM, Li D, Risch HA, Silverman DT, Ji BT, Gallinger S, Holly EA, Fontham EH, Maisonneuve P, Bueno-de-Mesquita HB, Ghadirian P, Kurtz RC, Ludwig E, Yu H, Lowenfels AB, Seminara D, Petersen GM, La Vecchia C, Boffetta P. Pancreatitis and pancreatic cancer risk: a pooled analysis in the International Pancreatic Cancer Case-Control Consortium (PanC4). Ann Oncol 2012; 23(11): 2964–2970

[16]

Cai J, Chen H, Lu M, Zhang Y, Lu B, You L, Zhang T, Dai M, Zhao Y. Advances in the epidemiology of pancreatic cancer: trends, risk factors, screening, and prognosis. Cancer Lett 2021; 520: 1–11

[17]

Bosetti C, Lucenteforte E, Silverman DT, Petersen G, Bracci PM, Ji BT, Negri E, Li D, Risch HA, Olson SH, Gallinger S, Miller AB, Bueno-de-Mesquita HB, Talamini R, Polesel J, Ghadirian P, Baghurst PA, Zatonski W, Fontham E, Bamlet WR, Holly EA, Bertuccio P, Gao YT, Hassan M, Yu H, Kurtz RC, Cotterchio M, Su J, Maisonneuve P, Duell EJ, Boffetta P, La Vecchia C. Cigarette smoking and pancreatic cancer: an analysis from the International Pancreatic Cancer Case-Control Consortium (Panc4). Ann Oncol 2012; 23(7): 1880–1888

[18]

Iodice S, Gandini S, Maisonneuve P, Lowenfels AB. Tobacco and the risk of pancreatic cancer: a review and meta-analysis. Langenbecks Arch Surg 2008; 393(4): 535–545

[19]

Sung H, Siegel RL, Rosenberg PS, Jemal A. Emerging cancer trends among young adults in the USA: analysis of a population-based cancer registry. Lancet Public Health 2019; 4(3): e137–e147

[20]

Elena JW, Steplowski E, Yu K, Hartge P, Tobias GS, Brotzman MJ, Chanock SJ, Stolzenberg-Solomon RZ, Arslan AA, Bueno-de-Mesquita HB, Helzlsouer K, Jacobs EJ, LaCroix A, Petersen G, Zheng W, Albanes D, Allen NE, Amundadottir L, Bao Y, Boeing H, Boutron-Ruault MC, Buring JE, Gaziano JM, Giovannucci EL, Duell EJ, Hallmans G, Howard BV, Hunter DJ, Hutchinson A, Jacobs KB, Kooperberg C, Kraft P, Mendelsohn JB, Michaud DS, Palli D, Phillips LS, Overvad K, Patel AV, Sansbury L, Shu XO, Simon MS, Slimani N, Trichopoulos D, Visvanathan K, Virtamo J, Wolpin BM, Zeleniuch-Jacquotte A, Fuchs CS, Hoover RN, Gross M. Diabetes and risk of pancreatic cancer: a pooled analysis from the pancreatic cancer cohort consortium. Cancer Causes Control 2013; 24(1): 13–25

[21]

Bosetti C, Rosato V, Li D, Silverman D, Petersen GM, Bracci PM, Neale RE, Muscat J, Anderson K, Gallinger S, Olson SH, Miller AB, Bas Bueno-de-Mesquita H, Scelo G, Janout V, Holcatova I, Lagiou P, Serraino D, Lucenteforte E, Fabianova E, Baghurst PA, Zatonski W, Foretova L, Fontham E, Bamlet WR, Holly EA, Negri E, Hassan M, Prizment A, Cotterchio M, Cleary S, Kurtz RC, Maisonneuve P, Trichopoulos D, Polesel J, Duell EJ, Boffetta P, La Vecchia C, Ghadirian P. Diabetes, antidiabetic medications, and pancreatic cancer risk: an analysis from the International Pancreatic Cancer Case-Control Consortium. Ann Oncol 2014; 25(10): 2065–2072

[22]

Genkinger JM, Spiegelman D, Anderson KE, Bergkvist L, Bernstein L, van den Brandt PA, English DR, Freudenheim JL, Fuchs CS, Giles GG, Giovannucci E, Hankinson SE, Horn-Ross PL, Leitzmann M, Männistö S, Marshall JR, McCullough ML, Miller AB, Reding DJ, Robien K, Rohan TE, Schatzkin A, Stevens VL, Stolzenberg-Solomon RZ, Verhage BA, Wolk A, Ziegler RG, Smith-Warner SA. Alcohol intake and pancreatic cancer risk: a pooled analysis of fourteen cohort studies. Cancer Epidemiol Biomarkers Prev 2009; 18(3): 765–776

[23]

Yadav D, Lowenfels AB. The epidemiology of pancreatitis and pancreatic cancer. Gastroenterology 2013; 144(6): 1252–1261

[24]

Xu JH, Fu JJ, Wang XL, Zhu JY, Ye XH, Chen SD. Hepatitis B or C viral infection and risk of pancreatic cancer: a meta-analysis of observational studies. World J Gastroenterol 2013; 19(26): 4234–4241

[25]

Kamiza AB, Su FH, Wang WC, Sung FC, Chang SN, Yeh CC. Chronic hepatitis infection is associated with extrahepatic cancer development: a nationwide population-based study in Taiwan. BMC Cancer 2016; 16(1): 861

[26]

Allison RD, Tong X, Moorman AC, Ly KN, Rupp L, Xu F, Gordon SC, Holmberg SD; Chronic Hepatitis Cohort Study (CHeCS) Investigators. Increased incidence of cancer and cancer-related mortality among persons with chronic hepatitis C infection, 2006–2010. J Hepatol 2015; 63(4): 822–828

[27]

Krull Abe S, Inoue M, Sawada N, Iwasaki M, Shimazu T, Yamaji T, Sasazuki S, Saito E, Tanaka Y, Mizokami M, Tsugane S; JPHC Study Group. Hepatitis B and C virus infection and risk of pancreatic cancer: a population-based cohort study (JPHC Study Cohort II). Cancer Epidemiol Biomarkers Prev 2016; 25(3): 555–557

[28]

Huang J, Zagai U, Hallmans G, Nyrén O, Engstrand L, Stolzenberg-Solomon R, Duell EJ, Overvad K, Katzke VA, Kaaks R, Jenab M, Park JY, Murillo R, Trichopoulou A, Lagiou P, Bamia C, Bradbury KE, Riboli E, Aune D, Tsilidis KK, Capellá G, Agudo A, Krogh V, Palli D, Panico S, Weiderpass E, Tjønneland A, Olsen A, Martínez B, Redondo-Sanchez D, Chirlaque MD, Hm Peeters P, Regnér S, Lindkvist B, Naccarati A, Ardanaz E, Larrañaga N, Boutron-Ruault MC, Rebours V, Barré A, Bueno-de-Mesquita HB, Ye W. Helicobacter pylori infection, chronic corpus atrophic gastritis and pancreatic cancer risk in the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort: a nested case-control study. Int J Cancer 2017; 140(8): 1727–1735

[29]

GBD 2017 Pancreatic Cancer Collaborators. The global, regional, and national burden of pancreatic cancer and its attributable risk factors in 195 countries and territories, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol Hepatol 2019; 4(12): 934–947

[30]

Zaitsu M, Kim Y, Lee HE, Takeuchi T, Kobayashi Y, Kawachi I. Occupational class differences in pancreatic cancer survival: a population-based cancer registry-based study in Japan. Cancer Med 2019; 8(6): 3261–3268

[31]

Fan X, Alekseyenko AV, Wu J, Peters BA, Jacobs EJ, Gapstur SM, Purdue MP, Abnet CC, Stolzenberg-Solomon R, Miller G, Ravel J, Hayes RB, Ahn J. Human oral microbiome and prospective risk for pancreatic cancer: a population-based nested case-control study. Gut 2018; 67(1): 120–127

[32]

Cotterchio M, Lowcock E, Hudson TJ, Greenwood C, Gallinger S. Association between allergies and risk of pancreatic cancer. Cancer Epidemiol Biomarkers Prev 2014; 23(3): 469–480

[33]

Singhi AD, Ishida H, Ali SZ, Goggins M, Canto M, Wolfgang C, Meriden Z, Roberts N, Klein AP, Hruban RH. A histomorphologic comparison of familial and sporadic pancreatic cancers. Pancreatology 2015; 15(4): 387–391

[34]

Overbeek KA, Levink IJM, Koopmann BDM, Harinck F, Konings ICAW, Ausems MGEM, Wagner A, Fockens P, van Eijck CH, Groot Koerkamp B, Busch ORC, Besselink MG, Bastiaansen BAJ, van Driel LMJW, Erler NS, Vleggaar FP, Poley JW, Cahen DL, van Hooft JE, Bruno MJ; Dutch Familial Pancreatic Cancer Surveillance Study Group. Long-term yield of pancreatic cancer surveillance in high-risk individuals. Gut 2022; 71(6): 1152–1160

[35]

Wu C, Miao X, Huang L, Che X, Jiang G, Yu D, Yang X, Cao G, Hu Z, Zhou Y, Zuo C, Wang C, Zhang X, Zhou Y, Yu X, Dai W, Li Z, Shen H, Liu L, Chen Y, Zhang S, Wang X, Zhai K, Chang J, Liu Y, Sun M, Cao W, Gao J, Ma Y, Zheng X, Cheung ST, Jia Y, Xu J, Tan W, Zhao P, Wu T, Wang C, Lin D. Genome-wide association study identifies five loci associated with susceptibility to pancreatic cancer in Chinese populations. Nat Genet 2012; 44: 62–66

[36]

Holter S, Borgida A, Dodd A, Grant R, Semotiuk K, Hedley D, Dhani N, Narod S, Akbari M, Moore M, Gallinger S. Germline BRCA mutations in a large clinic-based cohort of patients with pancreatic adenocarcinoma. J Clin Oncol 2015; 33(28): 3124–3129

[37]

Grant RC, Denroche RE, Borgida A, Virtanen C, Cook N, Smith AL, Connor AA, Wilson JM, Peterson G, Roberts NJ, Klein AP, Grimmond SM, Biankin A, Cleary S, Moore M, Lemire M, Zogopoulos G, Stein L, Gallinger S. Exome-wide association study of pancreatic cancer risk. Gastroenterology 2018; 154(3): 719–722.e3

[38]

Hu C, Hart SN, Polley EC, Gnanaolivu R, Shimelis H, Lee KY, Lilyquist J, Na J, Moore R, Antwi SO, Bamlet WR, Chaffee KG, DiCarlo J, Wu Z, Samara R, Kasi PM, McWilliams RR, Petersen GM, Couch FJ. Association between inherited germline mutations in cancer predisposition genes and risk of pancreatic cancer. JAMA 2018; 319(23): 2401–2409

[39]

Klein AP. Pancreatic cancer epidemiology: understanding the role of lifestyle and inherited risk factors. Nat Rev Gastroenterol Hepatol 2021; 18(7): 493–502

[40]

Rainone M, Singh I, Salo-Mullen EE, Stadler ZK, O’Reilly EM. An emerging paradigm for germline testing in pancreatic ductal adenocarcinoma and immediate implications for clinical practice: a review. JAMA Oncol 2020; 6(5): 764–771

[41]

Buscail L, Bournet B, Cordelier P. Role of oncogenic KRAS in the diagnosis, prognosis and treatment of pancreatic cancer. Nat Rev Gastroenterol Hepatol 2020; 17(3): 153–168

[42]

Singh K, Pruski M, Bland R, Younes M, Guha S, Thosani N, Maitra A, Cash BD, McAllister F, Logsdon CD, Chang JT, Bailey-Lundberg JM. Kras mutation rate precisely orchestrates ductal derived pancreatic intraepithelial neoplasia and pancreatic cancer. Lab Invest 2021; 101(2): 177–192

[43]

Cancer Genome Atlas Research Network. Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell 2017; 32(2): 185–203.e13

[44]

Witkiewicz AK, McMillan EA, Balaji U, Baek G, Lin WC, Mansour J, Mollaee M, Wagner KU, Koduru P, Yopp A, Choti MA, Yeo CJ, McCue P, White MA, Knudsen ES. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat Commun 2015; 6(1): 6744

[45]

O'Reilly EM, Hechtman JF. Tumour response to TRK inhibition in a patient with pancreatic adenocarcinoma harbouring an NTRK gene fusion. Ann Oncol 2019; 30(Suppl_8): viii36–viii40

[46]

Heining C, Horak P, Uhrig S, Codo PL, Klink B, Hutter B, Fröhlich M, Bonekamp D, Richter D, Steiger K, Penzel R, Endris V, Ehrenberg KR, Frank S, Kleinheinz K, Toprak UH, Schlesner M, Mandal R, Schulz L, Lambertz H, Fetscher S, Bitzer M, Malek NP, Horger M, Giese NA, Strobel O, Hackert T, Springfeld C, Feuerbach L, Bergmann F, Schröck E, von Kalle C, Weichert W, Scholl C, Ball CR, Stenzinger A, Brors B, Fröhling S, Glimm H. NRG1 fusions in KRAS wild-type pancreatic cancer. Cancer Discov 2018; 8(9): 1087–1095

[47]

Jones MR, Williamson LM, Topham JT, Lee MKC, Goytain A, Ho J, Denroche RE, Jang G, Pleasance E, Shen Y, Karasinska JM, McGhie JP, Gill S, Lim HJ, Moore MJ, Wong HL, Ng T, Yip S, Zhang W, Sadeghi S, Reisle C, Mungall AJ, Mungall KL, Moore RA, Ma Y, Knox JJ, Gallinger S, Laskin J, Marra MA, Schaeffer DF, Jones SJM, Renouf DJ. NRG1 gene fusions are recurrent, clinically actionable gene rearrangements in KRAS wild-type pancreatic ductal adenocarcinoma. Clin Cancer Res 2019; 25(15): 4674–4681

[48]

Hayashi A, Hong J, Iacobuzio-Donahue CA. The pancreatic cancer genome revisited. Nat Rev Gastroenterol Hepatol 2021; 18(7): 469–481

[49]

Connor AA, Denroche RE, Jang GH, Lemire M, Zhang A, Chan-Seng-Yue M, Wilson G, Grant RC, Merico D, Lungu I, Bartlett JMS, Chadwick D, Liang SB, Eagles J, Mbabaali F, Miller JK, Krzyzanowski P, Armstrong H, Luo X, Jorgensen LGT, Romero JM, Bavi P, Fischer SE, Serra S, Hafezi-Bakhtiari S, Caglar D, Roehrl MHA, Cleary S, Hollingsworth MA, Petersen GM, Thayer S, Law CHL, Nanji S, Golan T, Smith AL, Borgida A, Dodd A, Hedley D, Wouters BG, O’Kane GM, Wilson JM, Zogopoulos G, Notta F, Knox JJ, Gallinger S. Integration of genomic and transcriptional features in pancreatic cancer reveals increased cell cycle progression in metastases. Cancer Cell 2019; 35(2): 267–282.e7

[50]

Cao L, Huang C, Cui Zhou D, Hu Y, Lih TM, Savage SR, Krug K, Clark DJ, Schnaubelt M, Chen L, da Veiga Leprevost F, Eguez RV, Yang W, Pan J, Wen B, Dou Y, Jiang W, Liao Y, Shi Z, Terekhanova NV, Cao S, Lu RJ, Li Y, Liu R, Zhu H, Ronning P, Wu Y, Wyczalkowski MA, Easwaran H, Danilova L, Mer AS, Yoo S, Wang JM, Liu W, Haibe-Kains B, Thiagarajan M, Jewell SD, Hostetter G, Newton CJ, Li QK, Roehrl MH, Fenyö D, Wang P, Nesvizhskii AI, Mani DR, Omenn GS, Boja ES, Mesri M, Robles AI, Rodriguez H, Bathe OF, Chan DW, Hruban RH, Ding L, Zhang B, Zhang H; Clinical Proteomic Tumor Analysis Consortium. Proteogenomic characterization of pancreatic ductal adenocarcinoma. Cell 2021; 184(19): 5031–5052.e26

[51]

Xie D, Wang Z, Sun B, Qu L, Zeng M, Feng L, Guo M, Wang G, Hao J, Zhou G. High frequency of alternative splicing variants of the oncogene Focal Adhesion Kinase in neuroendocrine tumors of the pancreas and breast. Front Med 2023; 17(5): 907–923

[52]

Liu M. Arid1a: a gatekeeper in the development of pancreatic cancer from a rare precursor lesion. Gastroenterology 2022; 163(2): 371–373

[53]

Christenson ES, Jaffee E, Azad NS. Current and emerging therapies for patients with advanced pancreatic ductal adenocarcinoma: a bright future. Lancet Oncol 2020; 21(3): e135–e145

[54]

Ahmed S, Bradshaw AD, Gera S, Dewan MZ, Xu R. The TGF-β/Smad4 signaling pathway in pancreatic carcinogenesis and its clinical significance. J Clin Med 2017; 6(1): 5

[55]

Chan-Seng-Yue M, Kim JC, Wilson GW, Ng K, Figueroa EF, O’Kane GM, Connor AA, Denroche RE, Grant RC, McLeod J, Wilson JM, Jang GH, Zhang A, Dodd A, Liang SB, Borgida A, Chadwick D, Kalimuthu S, Lungu I, Bartlett JMS, Krzyzanowski PM, Sandhu V, Tiriac H, Froeling FEM, Karasinska JM, Topham JT, Renouf DJ, Schaeffer DF, Jones SJM, Marra MA, Laskin J, Chetty R, Stein LD, Zogopoulos G, Haibe-Kains B, Campbell PJ, Tuveson DA, Knox JJ, Fischer SE, Gallinger S, Notta F. Transcription phenotypes of pancreatic cancer are driven by genomic events during tumor evolution. Nat Genet 2020; 52(2): 231–240

[56]

Brunner M, Wu Z, Krautz C, Pilarsky C, Grützmann R, Weber GF. Current clinical strategies of pancreatic cancer treatment and open molecular questions. Int J Mol Sci 2019; 20(18): 4543

[57]

Gao J, Wang L, Xu J, Zheng J, Man X, Wu H, Jin J, Wang K, Xiao H, Li S, Li Z. Aberrant DNA methyltransferase expression in pancreatic ductal adenocarcinoma development and progression. J Exp Clin Cancer Res 2013; 32(1): 86

[58]

Zhang JJ, Zhu Y, Zhu Y, Wu JL, Liang WB, Zhu R, Xu ZK, Du Q, Miao Y. Association of increased DNA methyltransferase expression with carcinogenesis and poor prognosis in pancreatic ductal adenocarcinoma. Clin Transl Oncol 2012; 14(2): 116–124

[59]

Nagtegaal ID, Odze RD, Klimstra D, Paradis V, Rugge M, Schirmacher P, Washington KM, Carneiro F, Cree IA; WHO Classification of Tumours Editorial Board. The 2019 WHO classification of tumours of the digestive system. Histopathology 2020; 76(2): 182–188

[60]

Zhang L, Chen D, Song D, Liu X, Zhang Y, Xu X, Wang X. Clinical and translational values of spatial transcriptomics. Signal Transduct Target Ther 2022; 7(1): 111

[61]

Schäfer D, Tomiuk S, Küster LN, Rawashdeh WA, Henze J, Tischler-Höhle G, Agorku DJ, Brauner J, Linnartz C, Lock D, Kaiser A, Herbel C, Eckardt D, Lamorte M, Lenhard D, Schüler J, Ströbel P, Missbach-Guentner J, Pinkert-Leetsch D, Alves F, Bosio A, Hardt O. Identification of CD318, TSPAN8 and CD66c as target candidates for CAR T cell based immunotherapy of pancreatic adenocarcinoma. Nat Commun 2021; 12(1): 1453

[62]

Hsieh WC, Budiarto BR, Wang YF, Lin CY, Gwo MC, So DK, Tzeng YS, Chen SY. Spatial multi-omics analyses of the tumor immune microenvironment. J Biomed Sci 2022; 29(1): 96

[63]

Moncada R, Barkley D, Wagner F, Chiodin M, Devlin JC, Baron M, Hajdu CH, Simeone DM, Yanai I. Integrating microarray-based spatial transcriptomics and single-cell RNA-seq reveals tissue architecture in pancreatic ductal adenocarcinomas. Nat Biotechnol 2020; 38(3): 333–342

[64]

Hwang WL, Jagadeesh KA, Guo JA, Hoffman HI, Yadollahpour P, Reeves JW, Mohan R, Drokhlyansky E, Van Wittenberghe N, Ashenberg O, Farhi SL, Schapiro D, Divakar P, Miller E, Zollinger DR, Eng G, Schenkel JM, Su J, Shiau C, Yu P, Freed-Pastor WA, Abbondanza D, Mehta A, Gould J, Lambden C, Porter CBM, Tsankov A, Dionne D, Waldman J, Cuoco MS, Nguyen L, Delorey T, Phillips D, Barth JL, Kem M, Rodrigues C, Ciprani D, Roldan J, Zelga P, Jorgji V, Chen JH, Ely Z, Zhao D, Fuhrman K, Fropf R, Beechem JM, Loeffler JS, Ryan DP, Weekes CD, Ferrone CR, Qadan M, Aryee MJ, Jain RK, Neuberg DS, Wo JY, Hong TS, Xavier R, Aguirre AJ, Rozenblatt-Rosen O, Mino-Kenudson M, Castillo CF, Liss AS, Ting DT, Jacks T, Regev A. Single-nucleus and spatial transcriptome profiling of pancreatic cancer identifies multicellular dynamics associated with neoadjuvant treatment. Nat Genet 2022; 54(8): 1178–1191

[65]

Cui Zhou D, Jayasinghe RG, Chen S, Herndon JM, Iglesia MD, Navale P, Wendl MC, Caravan W, Sato K, Storrs E, Mo CK, Liu J, Southard-Smith AN, Wu Y, Naser Al Deen N, Baer JM, Fulton RS, Wyczalkowski MA, Liu R, Fronick CC, Fulton LA, Shinkle A, Thammavong L, Zhu H, Sun H, Wang LB, Li Y, Zuo C, McMichael JF, Davies SR, Appelbaum EL, Robbins KJ, Chasnoff SE, Yang X, Reeb AN, Oh C, Serasanambati M, Lal P, Varghese R, Mashl JR, Ponce J, Terekhanova NV, Yao L, Wang F, Chen L, Schnaubelt M, Lu RJ, Schwarz JK, Puram SV, Kim AH, Song SK, Shoghi KI, Lau KS, Ju T, Chen K, Chatterjee D, Hawkins WG, Zhang H, Achilefu S, Chheda MG, Oh ST, Gillanders WE, Chen F, DeNardo DG, Fields RC, Ding L. Spatially restricted drivers and transitional cell populations cooperate with the microenvironment in untreated and chemo-resistant pancreatic cancer. Nat Genet 2022; 54(9): 1390–1405

[66]

Tosti L, Hang Y, Debnath O, Tiesmeyer S, Trefzer T, Steiger K, Ten FW, Lukassen S, Ballke S, Kühl AA, Spieckermann S, Bottino R, Ishaque N, Weichert W, Kim SK, Eils R, Conrad C. Single-nucleus and in situ RNA-sequencing reveal cell topographies in the human pancreas. Gastroenterology 2021; 160(4): 1330–1344.e11

[67]

Barkley D, Moncada R, Pour M, Liberman DA, Dryg I, Werba G, Wang W, Baron M, Rao A, Xia B, França GS, Weil A, Delair DF, Hajdu C, Lund AW, Osman I, Yanai I. Cancer cell states recur across tumor types and form specific interactions with the tumor microenvironment. Nat Genet 2022; 54(8): 1192–1201

[68]

Hayashi A, Fan J, Chen R, Ho YJ, Makohon-Moore AP, Lecomte N, Zhong Y, Hong J, Huang J, Sakamoto H, Attiyeh MA, Kohutek ZA, Zhang L, Boumiza A, Kappagantula R, Baez P, Bai J, Lisi M, Chadalavada K, Melchor JP, Wong W, Nanjangud GJ, Basturk O, O’Reilly EM, Klimstra DS, Hruban RH, Wood LD, Overholtzer M, Iacobuzio-Donahue CA. A unifying paradigm for transcriptional heterogeneity and squamous features in pancreatic ductal adenocarcinoma. Nat Cancer 2020; 1(1): 59–74

[69]

Bailey P, Chang DK, Nones K, Johns AL, Patch AM, Gingras MC, Miller DK, Christ AN, Bruxner TJ, Quinn MC, Nourse C, Murtaugh LC, Harliwong I, Idrisoglu S, Manning S, Nourbakhsh E, Wani S, Fink L, Holmes O, Chin V, Anderson MJ, Kazakoff S, Leonard C, Newell F, Waddell N, Wood S, Xu Q, Wilson PJ, Cloonan N, Kassahn KS, Taylor D, Quek K, Robertson A, Pantano L, Mincarelli L, Sanchez LN, Evers L, Wu J, Pinese M, Cowley MJ, Jones MD, Colvin EK, Nagrial AM, Humphrey ES, Chantrill LA, Mawson A, Humphris J, Chou A, Pajic M, Scarlett CJ, Pinho AV, Giry-Laterriere M, Rooman I, Samra JS, Kench JG, Lovell JA, Merrett ND, Toon CW, Epari K, Nguyen NQ, Barbour A, Zeps N, Moran-Jones K, Jamieson NB, Graham JS, Duthie F, Oien K, Hair J, Grützmann R, Maitra A, Iacobuzio-Donahue CA, Wolfgang CL, Morgan RA, Lawlor RT, Corbo V, Bassi C, Rusev B, Capelli P, Salvia R, Tortora G, Mukhopadhyay D, Petersen GM; Australian Pancreatic Cancer Genome Initiative; Munzy DM, Fisher WE, Karim SA, Eshleman JR, Hruban RH, Pilarsky C, Morton JP, Sansom OJ, Scarpa A, Musgrove EA, Bailey UM, Hofmann O, Sutherland RL, Wheeler DA, Gill AJ, Gibbs RA, Pearson JV, Waddell N, Biankin AV, Grimmond SM. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 2016; 531(7592): 47–52

[70]

Sun H, Zhang D, Huang C, Guo Y, Yang Z, Yao N, Dong X, Cheng R, Zhao N, Meng J, Sun B, Hao J. Hypoxic microenvironment induced spatial transcriptome changes in pancreatic cancer. Cancer Biol Med 2021; 18(2): 616–630

[71]

BellATF. PanIN and CAF transitions in pancreatic carcinogenesis revealed with spatial data integration. bioRxiv 2022; 2022.07.16.500312

[72]

AgostiniA. Transcriptomic dissection of intraepithelial papillary mucinous neoplasms progression by spatial technologies identified novel markers of pancreatic carcinogenesis. bioRxiv 2022; 2022.10.12.511894

[73]

Sans M, Makino Y, Min J, Rajapakshe KI, Yip-Schneider M, Schmidt CM, Hurd MW, Burks JK, Gomez JA, Thege FI, Fahrmann JF, Wolff RA, Kim MP, Guerrero PA, Maitra A. Spatial transcriptomics of intraductal papillary mucinous neoplasms of the pancreas identifies NKX6-2 as a driver of gastric differentiation and indolent biological potential. Cancer Discov 2023; 13(8): 1844–1861

[74]

Peng J, Sun BF, Chen CY, Zhou JY, Chen YS, Chen H, Liu L, Huang D, Jiang J, Cui GS, Yang Y, Wang W, Guo D, Dai M, Guo J, Zhang T, Liao Q, Liu Y, Zhao YL, Han DL, Zhao Y, Yang YG, Wu W. Single-cell RNA-seq highlights intra-tumoral heterogeneity and malignant progression in pancreatic ductal adenocarcinoma. Cell Res 2019; 29(9): 725–738

[75]

Ma Z, Lytle NK, Chen B, Jyotsana N, Novak SW, Cho CJ, Caplan L, Ben-Levy O, Neininger AC, Burnette DT, Trinh VQ, Tan MCB, Patterson EA, Arrojo E Drigo R, Giraddi RR, Ramos C, Means AL, Matsumoto I, Manor U, Mills JC, Goldenring JR, Lau KS, Wahl GM, DelGiorno KE. Single-cell transcriptomics reveals a conserved metaplasia program in pancreatic injury. Gastroenterology 2022; 162(2): 604–620.e20

[76]

Lee JJ, Bernard V, Semaan A, Monberg ME, Huang J, Stephens BM, Lin D, Rajapakshe KI, Weston BR, Bhutani MS, Haymaker CL, Bernatchez C, Taniguchi CM, Maitra A, Guerrero PA. Elucidation of tumor-stromal heterogeneity and the ligand-receptor interactome by single-cell transcriptomics in real-world pancreatic cancer biopsies. Clin Cancer Res 2021; 27(21): 5912–5921

[77]

Lin W, Noel P, Borazanci EH, Lee J, Amini A, Han IW, Heo JS, Jameson GS, Fraser C, Steinbach M, Woo Y, Fong Y, Cridebring D, Von Hoff DD, Park JO, Han H. Single-cell transcriptome analysis of tumor and stromal compartments of pancreatic ductal adenocarcinoma primary tumors and metastatic lesions. Genome Med 2020; 12(1): 80

[78]

Ligorio M, Sil S, Malagon-Lopez J, Nieman LT, Misale S, Di Pilato M, Ebright RY, Karabacak MN, Kulkarni AS, Liu A, Vincent Jordan N, Franses JW, Philipp J, Kreuzer J, Desai N, Arora KS, Rajurkar M, Horwitz E, Neyaz A, Tai E, Magnus NKC, Vo KD, Yashaswini CN, Marangoni F, Boukhali M, Fatherree JP, Damon LJ, Xega K, Desai R, Choz M, Bersani F, Langenbucher A, Thapar V, Morris R, Wellner UF, Schilling O, Lawrence MS, Liss AS, Rivera MN, Deshpande V, Benes CH, Maheswaran S, Haber DA, Fernandez-Del-Castillo C, Ferrone CR, Haas W, Aryee MJ, Ting DT. Stromal microenvironment shapes the intratumoral architecture of pancreatic cancer. Cell 2019; 178(1): 160–175.e27

[79]

Elyada E, Bolisetty M, Laise P, Flynn WF, Courtois ET, Burkhart RA, Teinor JA, Belleau P, Biffi G, Lucito MS, Sivajothi S, Armstrong TD, Engle DD, Yu KH, Hao Y, Wolfgang CL, Park Y, Preall J, Jaffee EM, Califano A, Robson P, Tuveson DA. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discov 2019; 9(8): 1102–1123

[80]

Foster DS, Januszyk M, Delitto D, Yost KE, Griffin M, Guo J, Guardino N, Delitto AE, Chinta M, Burcham AR, Nguyen AT, Bauer-Rowe KE, Titan AL, Salhotra A, Jones RE, da Silva O, Lindsay HG, Berry CE, Chen K, Henn D, Mascharak S, Talbott HE, Kim A, Nosrati F, Sivaraj D, Ransom RC, Matthews M, Khan A, Wagh D, Coller J, Gurtner GC, Wan DC, Wapnir IL, Chang HY, Norton JA, Longaker MT. Multiomic analysis reveals conservation of cancer-associated fibroblast phenotypes across species and tissue of origin. Cancer Cell 2022; 40(11): 1392–1406.e7

[81]

Dominguez CX, Müller S, Keerthivasan S, Koeppen H, Hung J, Gierke S, Breart B, Foreman O, Bainbridge TW, Castiglioni A, Senbabaoglu Y, Modrusan Z, Liang Y, Junttila MR, Klijn C, Bourgon R, Turley SJ. Single-cell RNA sequencing reveals stromal evolution into LRRC15+ myofibroblasts as a determinant of patient response to cancer immunotherapy. Cancer Discov 2020; 10(2): 232–253

[82]

Hutton C, Heider F, Blanco-Gomez A, Banyard A, Kononov A, Zhang X, Karim S, Paulus-Hock V, Watt D, Steele N, Kemp S, Hogg EKJ, Kelly J, Jackstadt RF, Lopes F, Menotti M, Chisholm L, Lamarca A, Valle J, Sansom OJ, Springer C, Malliri A, Marais R, Pasca di Magliano M, Zelenay S, Morton JP, Jørgensen C. Single-cell analysis defines a pancreatic fibroblast lineage that supports anti-tumor immunity. Cancer Cell 2021; 39(9): 1227–1244.e20

[83]

Hosein AN, Huang H, Wang Z, Parmar K, Du W, Huang J, Maitra A, Olson E, Verma U, Brekken RA. Cellular heterogeneity during mouse pancreatic ductal adenocarcinoma progression at single-cell resolution. JCI Insight 2019; 4(16): e129212

[84]

Sherman MH, Beatty GL. Tumor microenvironment in pancreatic cancer pathogenesis and therapeutic resistance. Annu Rev Pathol 2023; 18(1): 123–148

[85]

Steele NG, Carpenter ES, Kemp SB, Sirihorachai VR, The S, Delrosario L, Lazarus J, Amir ED, Gunchick V, Espinoza C, Bell S, Harris L, Lima F, Irizarry-Negron V, Paglia D, Macchia J, Chu AKY, Schofield H, Wamsteker EJ, Kwon R, Schulman A, Prabhu A, Law R, Sondhi A, Yu J, Patel A, Donahue K, Nathan H, Cho C, Anderson MA, Sahai V, Lyssiotis CA, Zou W, Allen BL, Rao A, Crawford HC, Bednar F, Frankel TL, Pasca di Magliano M. Multimodal mapping of the tumor and peripheral blood immune landscape in human pancreatic cancer. Nat Cancer 2020; 1(11): 1097–1112

[86]

Ho WJ, Erbe R, Danilova L, Phyo Z, Bigelow E, Stein-O’Brien G, Thomas DL 2nd, Charmsaz S, Gross N, Woolman S, Cruz K, Munday RM, Zaidi N, Armstrong TD, Sztein MB, Yarchoan M, Thompson ED, Jaffee EM, Fertig EJ. Multi-omic profiling of lung and liver tumor microenvironments of metastatic pancreatic cancer reveals site-specific immune regulatory pathways. Genome Biol 2021; 22(1): 154

[87]

Du Y, Cai Y, Lv Y, Zhang L, Yang H, Liu Q, Hong M, Teng Y, Tang W, Ma R, Wu J, Wu J, Wang Q, Chen H, Li K, Feng J. Single-cell RNA sequencing unveils the communications between malignant T and myeloid cells contributing to tumor growth and immunosuppression in cutaneous T-cell lymphoma. Cancer Lett 2022; 551: 215972

[88]

Shiau C, Su J, Guo JA, Hong TS, Wo JY, Jagadeesh KA, Hwang WL. Treatment-associated remodeling of the pancreatic cancer endothelium at single-cell resolution. Front Oncol 2022; 12: 929950

[89]

Du Y, Gu Z, Li Z, Yuan Z, Zhao Y, Zheng X, Bo X, Chen H, Wang C. Dynamic interplay between structural variations and 3D genome organization in pancreatic cancer. Adv Sci (Weinh) 2022; 9(18): e2200818

[90]

Alonso-Curbelo D, Ho YJ, Burdziak C, Maag JLV, Morris JP 4th, Chandwani R, Chen HA, Tsanov KM, Barriga FM, Luan W, Tasdemir N, Livshits G, Azizi E, Chun J, Wilkinson JE, Mazutis L, Leach SD, Koche R, Pe’er D, Lowe SW. A gene-environment-induced epigenetic program initiates tumorigenesis. Nature 2021; 590(7847): 642–648

[91]

Burdziak C, Alonso-Curbelo D, Walle T, Reyes J, Barriga FM, Haviv D, Xie Y, Zhao Z, Zhao CJ, Chen HA, Chaudhary O, Masilionis I, Choo ZN, Gao V, Luan W, Wuest A, Ho YJ, Wei Y, Quail DF, Koche R, Mazutis L, Chaligné R, Nawy T, Lowe SW, Pe’er D. Epigenetic plasticity cooperates with cell-cell interactions to direct pancreatic tumorigenesis. Science 2023; 380(6645): eadd5327

[92]

Guo F, Li L, Li J, Wu X, Hu B, Zhu P, Wen L, Tang F. Single-cell multi-omics sequencing of mouse early embryos and embryonic stem cells. Cell Res 2017; 27(8): 967–988

[93]

Fan X, Lu P, Wang H, Bian S, Wu X, Zhang Y, Liu Y, Fu D, Wen L, Hao J, Tang F. Integrated single-cell multiomics analysis reveals novel candidate markers for prognosis in human pancreatic ductal adenocarcinoma. Cell Discov 2022; 8(1): 13

[94]

Waddell N, Pajic M, Patch AM, Chang DK, Kassahn KS, Bailey P, Johns AL, Miller D, Nones K, Quek K, Quinn MC, Robertson AJ, Fadlullah MZ, Bruxner TJ, Christ AN, Harliwong I, Idrisoglu S, Manning S, Nourse C, Nourbakhsh E, Wani S, Wilson PJ, Markham E, Cloonan N, Anderson MJ, Fink JL, Holmes O, Kazakoff SH, Leonard C, Newell F, Poudel B, Song S, Taylor D, Waddell N, Wood S, Xu Q, Wu J, Pinese M, Cowley MJ, Lee HC, Jones MD, Nagrial AM, Humphris J, Chantrill LA, Chin V, Steinmann AM, Mawson A, Humphrey ES, Colvin EK, Chou A, Scarlett CJ, Pinho AV, Giry-Laterriere M, Rooman I, Samra JS, Kench JG, Pettitt JA, Merrett ND, Toon C, Epari K, Nguyen NQ, Barbour A, Zeps N, Jamieson NB, Graham JS, Niclou SP, Bjerkvig R, Grützmann R, Aust D, Hruban RH, Maitra A, Iacobuzio-Donahue CA, Wolfgang CL, Morgan RA, Lawlor RT, Corbo V, Bassi C, Falconi M, Zamboni G, Tortora G, Tempero MA; Australian Pancreatic Cancer Genome Initiative; Gill AJ, Eshleman JR, Pilarsky C, Scarpa A, Musgrove EA, Pearson JV, Biankin AV, Grimmond SM. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 2015; 518(7540): 495–501

[95]

Moffitt RA, Marayati R, Flate EL, Volmar KE, Loeza SG, Hoadley KA, Rashid NU, Williams LA, Eaton SC, Chung AH, Smyla JK, Anderson JM, Kim HJ, Bentrem DJ, Talamonti MS, Iacobuzio-Donahue CA, Hollingsworth MA, Yeh JJ. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat Genet 2015; 47(10): 1168–1178

[96]

Puleo F, Nicolle R, Blum Y, Cros J, Marisa L, Demetter P, Quertinmont E, Svrcek M, Elarouci N, Iovanna J, Franchimont D, Verset L, Galdon MG, Devière J, de Reyniès A, Laurent-Puig P, Van Laethem JL, Bachet JB, Maréchal R. Stratification of pancreatic ductal adenocarcinomas based on tumor and microenvironment features. Gastroenterology 2018; 155(6): 1999–2013.e3

[97]

Collisson EA, Sadanandam A, Olson P, Gibb WJ, Truitt M, Gu S, Cooc J, Weinkle J, Kim GE, Jakkula L, Feiler HS, Ko AH, Olshen AB, Danenberg KL, Tempero MA, Spellman PT, Hanahan D, Gray JW. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat Med 2011; 17(4): 500–503

[98]

Maurer C, Holmstrom SR, He J, Laise P, Su T, Ahmed A, Hibshoosh H, Chabot JA, Oberstein PE, Sepulveda AR, Genkinger JM, Zhang J, Iuga AC, Bansal M, Califano A, Olive KP. Experimental microdissection enables functional harmonisation of pancreatic cancer subtypes. Gut 2019; 68(6): 1034–1043

[99]

Jamal-Hanjani M, Wilson GA, McGranahan N, Birkbak NJ, Watkins TBK, Veeriah S, Shafi S, Johnson DH, Mitter R, Rosenthal R, Salm M, Horswell S, Escudero M, Matthews N, Rowan A, Chambers T, Moore DA, Turajlic S, Xu H, Lee SM, Forster MD, Ahmad T, Hiley CT, Abbosh C, Falzon M, Borg E, Marafioti T, Lawrence D, Hayward M, Kolvekar S, Panagiotopoulos N, Janes SM, Thakrar R, Ahmed A, Blackhall F, Summers Y, Shah R, Joseph L, Quinn AM, Crosbie PA, Naidu B, Middleton G, Langman G, Trotter S, Nicolson M, Remmen H, Kerr K, Chetty M, Gomersall L, Fennell DA, Nakas A, Rathinam S, Anand G, Khan S, Russell P, Ezhil V, Ismail B, Irvin-Sellers M, Prakash V, Lester JF, Kornaszewska M, Attanoos R, Adams H, Davies H, Dentro S, Taniere P, O’Sullivan B, Lowe HL, Hartley JA, Iles N, Bell H, Ngai Y, Shaw JA, Herrero J, Szallasi Z, Schwarz RF, Stewart A, Quezada SA, Le Quesne J, Van Loo P, Dive C, Hackshaw A, Swanton C; TRACERx Consortium. Tracking the evolution of non-small-cell lung cancer. N Engl J Med 2017; 376(22): 2109–2121

[100]

Rashid NU, Peng XL, Jin C, Moffitt RA, Volmar KE, Belt BA, Panni RZ, Nywening TM, Herrera SG, Moore KJ, Hennessey SG, Morrison AB, Kawalerski R, Nayyar A, Chang AE, Schmidt B, Kim HJ, Linehan DC, Yeh JJ. Purity independent subtyping of tumors (PurIST), a clinically robust, single-sample classifier for tumor subtyping in pancreatic cancer. Clin Cancer Res 2020; 26(1): 82–92

[101]

Ullman NA, Burchard PR, Dunne RF, Linehan DC. Immunologic strategies in pancreatic cancer: making cold tumors hot. J Clin Oncol 2022; 40(24): 2789–2805

[102]

Li X, Gulati M, Larson AC, Solheim JC, Jain M, Kumar S, Batra SK. Immune checkpoint blockade in pancreatic cancer: trudging through the immune desert. Semin Cancer Biol 2022; 86(Pt 2): 14–27

[103]

Nagaraju GP, Malla RR, Basha R, Motofei IG. Contemporary clinical trials in pancreatic cancer immunotherapy targeting PD-1 and PD-L1. Semin Cancer Biol 2022; 86(Pt 3): 616–621

[104]

Xue R, Zhang Q, Cao Q, Kong R, Xiang X, Liu H, Feng M, Wang F, Cheng J, Li Z, Zhan Q, Deng M, Zhu J, Zhang Z, Zhang N. Liver tumour immune microenvironment subtypes and neutrophil heterogeneity. Nature 2022; 612(7938): 141–147

[105]

Raghavan S, Winter PS, Navia AW, Williams HL, DenAdel A, Lowder KE, Galvez-Reyes J, Kalekar RL, Mulugeta N, Kapner KS, Raghavan MS, Borah AA, Liu N, Väyrynen SA, Costa AD, Ng RWS, Wang J, Hill EK, Ragon DY, Brais LK, Jaeger AM, Spurr LF, Li YY, Cherniack AD, Booker MA, Cohen EF, Tolstorukov MY, Wakiro I, Rotem A, Johnson BE, McFarland JM, Sicinska ET, Jacks TE, Sullivan RJ, Shapiro GI, Clancy TE, Perez K, Rubinson DA, Ng K, Cleary JM, Crawford L, Manalis SR, Nowak JA, Wolpin BM, Hahn WC, Aguirre AJ, Shalek AK. Microenvironment drives cell state, plasticity, and drug response in pancreatic cancer. Cell 2021; 184(25): 6119–6137.e26

[106]

Grimont A, Leach SD, Chandwani R. Uncertain beginnings: acinar and ductal cell plasticity in the development of pancreatic cancer. Cell Mol Gastroenterol Hepatol 2022; 13(2): 369–382

[107]

Messal HA, Alt S, Ferreira RMM, Gribben C, Wang VM, Cotoi CG, Salbreux G, Behrens A. Tissue curvature and apicobasal mechanical tension imbalance instruct cancer morphogenesis. Nature 2019; 566(7742): 126–130

[108]

Li S, Xie K. Ductal metaplasia in pancreas. Biochim Biophys Acta Rev Cancer 2022; 1877(2): 188698

[109]

Parte S, Nimmakayala RK, Batra SK, Ponnusamy MP. Acinar to ductal cell trans-differentiation: a prelude to dysplasia and pancreatic ductal adenocarcinoma. Biochim Biophys Acta Rev Cancer 2022; 1877(1): 188669

[110]

Del Poggetto E, Ho IL, Balestrieri C, Yen EY, Zhang S, Citron F, Shah R, Corti D, Diaferia GR, Li CY, Loponte S, Carbone F, Hayakawa Y, Valenti G, Jiang S, Sapio L, Jiang H, Dey P, Gao S, Deem AK, Rose-John S, Yao W, Ying H, Rhim AD, Genovese G, Heffernan TP, Maitra A, Wang TC, Wang L, Draetta GF, Carugo A, Natoli G, Viale A. Epithelial memory of inflammation limits tissue damage while promoting pancreatic tumorigenesis. Science 2021; 373(6561): eabj0486

[111]

De La O JP, Emerson LL, Goodman JL, Froebe SC, Illum BE, Curtis AB, Murtaugh LC. Notch and Kras reprogram pancreatic acinar cells to ductal intraepithelial neoplasia. Proc Natl Acad Sci USA 2008; 105(48): 18907–18912

[112]

Huang H, He M, Zhang Y, Zhang B, Niu Z, Zheng Y, Li W, Cui P, Wang X, Sun Q. Identification and validation of heterotypic cell-in-cell structure as an adverse prognostic predictor for young patients of resectable pancreatic ductal adenocarcinoma. Signal Transduct Target Ther 2020; 5(1): 246

[113]

Song J, Ruze R, Chen Y, Xu R, Yin X, Wang C, Xu Q, Zhao Y. Construction of a novel model based on cell-in-cell-related genes and validation of KRT7 as a biomarker for predicting survival and immune microenvironment in pancreatic cancer. BMC Cancer 2022; 22(1): 894

[114]

Xi NM, Li JJ. Benchmarking computational doublet-detection methods for single-cell RNA sequencing data. Cell Syst 2021; 12(2): 176–194.e6

[115]

Bais AS, Kostka D. scds: computational annotation of doublets in single-cell RNA sequencing data. Bioinformatics 2020; 36(4): 1150–1158

[116]

Singh VK, Yadav D, Garg PK. Diagnosis and management of chronic pancreatitis: a review. JAMA 2019; 322(24): 2422–2434

[117]

Greenhalf W, Lévy P, Gress T, Rebours V, Brand RE, Pandol S, Chari S, Jørgensen MT, Mayerle J, Lerch MM, Hegyi P, Kleeff J, Castillo CF, Isaji S, Shimosegawa T, Sheel A, Halloran CM, Garg P, Takaori K, Besselink MG, Forsmark CE, Wilcox CM, Maisonneuve P, Yadav D, Whitcomb D, Neoptolemos J; Working group for the International (IAP – APA – JPS – EPC) Consensus Guidelines for Chronic Pancreatitis. International consensus guidelines on surveillance for pancreatic cancer in chronic pancreatitis. Recommendations from the working group for the international consensus guidelines for chronic pancreatitis in collaboration with the International Association of Pancreatology, the American Pancreatic Association, the Japan Pancreas Society, and European Pancreatic Club. Pancreatology 2020; 20(5): 910–918

[118]

Hegyi P, Párniczky A, Lerch MM, Sheel ARG, Rebours V, Forsmark CE, Del Chiaro M, Rosendahl J, de-Madaria E, Szücs Á, Takaori K, Yadav D, Gheorghe C, Rakonczay Z Jr, Molero X, Inui K, Masamune A, Fernandez-Del Castillo C, Shimosegawa T, Neoptolemos JP, Whitcomb DC, Sahin-Tóth M; Working Group for the International (IAP–APA–JPS–EPC) Consensus Guidelines for Chronic Pancreatitis. International consensus guidelines for risk factors in chronic pancreatitis. Recommendations from the working group for the international consensus guidelines for chronic pancreatitis in collaboration with the International Association of Pancreatology, the American Pancreatic Association, the Japan Pancreas Society, and European Pancreatic Club. Pancreatology 2020; 20(4): 579–585

[119]

Sun C, Liu M, An W, Mao X, Jiang H, Zou W, Wu H, Liao Z, Li Z. Heterozygous Spink1 c.194+2T>C mutant mice spontaneously develop chronic pancreatitis. Gut 2020; 69(5): 967–968

[120]

Geisz A, Sahin-Tóth M. A preclinical model of chronic pancreatitis driven by trypsinogen autoactivation. Nat Commun 2018; 9(1): 5033

[121]

Hegyi E, Sahin-Tóth M. Human CPA1 mutation causes digestive enzyme misfolding and chronic pancreatitis in mice. Gut 2019; 68(2): 301–312

[122]

Kichler A, Jang S. Chronic pancreatitis: epidemiology, diagnosis, and management updates. Drugs 2020; 80(12): 1155–1168

[123]

Storz P. Acinar cell plasticity and development of pancreatic ductal adenocarcinoma. Nat Rev Gastroenterol Hepatol 2017; 14(5): 296–304

[124]

Westphalen CB, Takemoto Y, Tanaka T, Macchini M, Jiang Z, Renz BW, Chen X, Ormanns S, Nagar K, Tailor Y, May R, Cho Y, Asfaha S, Worthley DL, Hayakawa Y, Urbanska AM, Quante M, Reichert M, Broyde J, Subramaniam PS, Remotti H, Su GH, Rustgi AK, Friedman RA, Honig B, Califano A, Houchen CW, Olive KP, Wang TC. Dclk1 defines quiescent pancreatic progenitors that promote injury-induced regeneration and tumorigenesis. Cell Stem Cell 2016; 18(4): 441–455

[125]

Bailey JM, Alsina J, Rasheed ZA, McAllister FM, Fu YY, Plentz R, Zhang H, Pasricha PJ, Bardeesy N, Matsui W, Maitra A, Leach SD. DCLK1 marks a morphologically distinct subpopulation of cells with stem cell properties in preinvasive pancreatic cancer. Gastroenterology 2014; 146(1): 245–256

[126]

Ferguson FM, Nabet B, Raghavan S, Liu Y, Leggett AL, Kuljanin M, Kalekar RL, Yang A, He S, Wang J, Ng RWS, Sulahian R, Li L, Poulin EJ, Huang L, Koren J, Dieguez-Martinez N, Espinosa S, Zeng Z, Corona CR, Vasta JD, Ohi R, Sim T, Kim ND, Harshbarger W, Lizcano JM, Robers MB, Muthaswamy S, Lin CY, Look AT, Haigis KM, Mancias JD, Wolpin BM, Aguirre AJ, Hahn WC, Westover KD, Gray NS. Discovery of a selective inhibitor of doublecortin like kinase 1. Nat Chem Biol 2020; 16(6): 635–643

[127]

Basturk O, Hong SM, Wood LD, Adsay NV, Albores-Saavedra J, Biankin AV, Brosens LA, Fukushima N, Goggins M, Hruban RH, Kato Y, Klimstra DS, Klöppel G, Krasinskas A, Longnecker DS, Matthaei H, Offerhaus GJ, Shimizu M, Takaori K, Terris B, Yachida S, Esposito I, Furukawa T; Baltimore Consensus Meeting. A revised classification system and recommendations from the Baltimore Consensus Meeting for Neoplastic Precursor Lesions in the Pancreas. Am J Surg Pathol 2015; 39(12): 1730–1741

[128]

Liffers ST, Godfrey L, Frohn L, Haeberle L, Yavas A, Vesce R, Goering W, Opitz FV, Stoecklein N, Knoefel WT, Schlitter AM, Klöppel G, Espinet E, Trumpp A, Siveke JT, Esposito I. Molecular heterogeneity and commonalities in pancreatic cancer precursors with gastric and intestinal phenotype. Gut 2023; 72(3): 522–534

[129]

Felsenstein M, Noë M, Masica DL, Hosoda W, Chianchiano P, Fischer CG, Lionheart G, Brosens LAA, Pea A, Yu J, Gemenetzis G, Groot VP, Makary MA, He J, Weiss MJ, Cameron JL, Wolfgang CL, Hruban RH, Roberts NJ, Karchin R, Goggins MG, Wood LD. IPMNs with co-occurring invasive cancers: neighbours but not always relatives. Gut 2018; 67(9): 1652–1662

[130]

Scarpa A, Real FX, Luchini C. Genetic unrelatedness of co-occurring pancreatic adenocarcinomas and IPMNs challenges current views of clinical management. Gut 2018; 67(9): 1561–1563

[131]

Mafficini A, Simbolo M, Shibata T, Hong SM, Pea A, Brosens LA, Cheng L, Antonello D, Sciammarella C, Cantù C, Mattiolo P, Taormina SV, Malleo G, Marchegiani G, Sereni E, Corbo V, Paolino G, Ciaparrone C, Hiraoka N, Pallaoro D, Jansen C, Milella M, Salvia R, Lawlor RT, Adsay V, Scarpa A, Luchini C. Integrative characterization of intraductal tubulopapillary neoplasm (ITPN) of the pancreas and associated invasive adenocarcinoma. Mod Pathol 2022; 35(12): 1929–1943

[132]

Yamaguchi H, Shimizu M, Ban S, Koyama I, Hatori T, Fujita I, Yamamoto M, Kawamura S, Kobayashi M, Ishida K, Morikawa T, Motoi F, Unno M, Kanno A, Satoh K, Shimosegawa T, Orikasa H, Watanabe T, Nishimura K, Ebihara Y, Koike N, Furukawa T. Intraductal tubulopapillary neoplasms of the pancreas distinct from pancreatic intraepithelial neoplasia and intraductal papillary mucinous neoplasms. Am J Surg Pathol 2009; 33(8): 1164–1172

[133]

Paolino G, Esposito I, Hong SM, Basturk O, Mattiolo P, Kaneko T, Veronese N, Scarpa A, Adsay V, Luchini C. Intraductal tubulopapillary neoplasm (ITPN) of the pancreas: a distinct entity among pancreatic tumors. Histopathology 2022; 81(3): 297–309

[134]

Fukunaga Y, Fukuda A, Omatsu M, Namikawa M, Sono M, Masuda T, Araki O, Nagao M, Yoshikawa T, Ogawa S, Hiramatsu Y, Muta Y, Tsuda M, Maruno T, Nakanishi Y, Ferrer J, Tsuruyama T, Masui T, Hatano E, Seno H. Loss of Arid1a and Pten in pancreatic ductal cells induces intraductal tubulopapillary neoplasm via the YAP/TAZ pathway. Gastroenterology 2022; 163(2): 466–480.e6

[135]

Basturk O, Berger MF, Yamaguchi H, Adsay V, Askan G, Bhanot UK, Zehir A, Carneiro F, Hong SM, Zamboni G, Dikoglu E, Jobanputra V, Wrzeszczynski KO, Balci S, Allen P, Ikari N, Takeuchi S, Akagawa H, Kanno A, Shimosegawa T, Morikawa T, Motoi F, Unno M, Higuchi R, Yamamoto M, Shimizu K, Furukawa T, Klimstra DS. Pancreatic intraductal tubulopapillary neoplasm is genetically distinct from intraductal papillary mucinous neoplasm and ductal adenocarcinoma. Mod Pathol 2017; 30(12): 1760–1772

[136]

Sakihama K, Koga Y, Yamamoto T, Shimada Y, Yamada Y, Kawat J, Shindo K, Nakamura M, Oda Y. RNF43 as a predictor of malignant transformation of pancreatic mucinous cystic neoplasm. Virchows Arch 2022; 480(6): 1189–1199

[137]

Conner JR, Enríquez AM, Kenudso MM, Garcia E, Pitman MB, Sholl LM, Srivastava A, Doyle LA. Genomic characterization of low- and high-grade pancreatic mucinous cystic neoplasms reveals recurrent kras alterations in “high-risk” lesions. Pancreas 2017; 46(5): 665–671

[138]

Maimaitiaili Y, Fukumura Y, Hirabayashi K, Kinowaki Y, Naito Y, Saito A, Rong L, Nakahodo J, Yao T. Investigation of -PRKACA/-PRKACB fusion genes in oncocytic tumors of the pancreatobiliary and other systems. Virchows Arch 2022; 481(6): 865–876

[139]

Singhi AD, Wood LD, Parks E, Torbenson MS, Felsenstein M, Hruban RH, Nikiforova MN, Wald AI, Kaya C, Nikiforov YE, Favazza L, He J, McGrath K, Fasanella KE, Brand RE, Lennon AM, Furlan A, Dasyam AK, Zureikat AH, Zeh HJ, Lee K, Bartlett DL, Slivka A. Recurrent rearrangements in PRKACA and PRKACB in intraductal oncocytic papillary neoplasms of the pancreas and bile duct. Gastroenterology 2020; 158(3): 573–582.e2

[140]

Wang T, Askan G, Adsay V, Allen P, Jarnagin WR, Memis B, Sigel C, Seven IE, Klimstra DS, Basturk O. Intraductal oncocytic papillary neoplasms: clinical-pathologic characterization of 24 cases, with an emphasis on associated invasive carcinomas. Am J Surg Pathol 2019; 43(5): 656–661

[141]

Ferreira RMM, Sancho R, Messal HA, Nye E, Spencer-Dene B, Stone RK, Stamp G, Rosewell I, Quaglia A, Behrens A. Duct- and acinar-derived pancreatic ductal adenocarcinomas show distinct tumor progression and marker expression. Cell Rep 2017; 21(4): 966–978

[142]

Makohon-Moore AP, Matsukuma K, Zhang M, Reiter JG, Gerold JM, Jiao Y, Sikkema L, Attiyeh MA, Yachida S, Sandone C, Hruban RH, Klimstra DS, Papadopoulos N, Nowak MA, Kinzler KW, Vogelstein B, Iacobuzio-Donahue CA. Precancerous neoplastic cells can move through the pancreatic ductal system. Nature 2018; 561(7722): 201–205

[143]

Hutchings D, Waters KM, Weiss MJ, Wolfgang CL, Makary MA, He J, Cameron JL, Wood LD, Hruban RH. Cancerization of the pancreatic ducts: demonstration of a common and under-recognized process using immunolabeling of paired duct lesions and invasive pancreatic ductal adenocarcinoma for p53 and Smad4 expression. Am J Surg Pathol 2018; 42(11): 1556–1561

[144]

Matsuda Y, Furukawa T, Yachida S, Nishimura M, Seki A, Nonaka K, Aida J, Takubo K, Ishiwata T, Kimura W, Arai T, Mino-Kenudson M. The prevalence and clinicopathological characteristics of high-grade pancreatic intraepithelial neoplasia: autopsy study evaluating the entire pancreatic parenchyma. Pancreas 2017; 46(5): 658–664

[145]

Goggins M, Hruban RH, Kern SE. BRCA2 is inactivated late in the development of pancreatic intraepithelial neoplasia: evidence and implications. Am J Pathol 2000; 156(5): 1767–1771

[146]

Sharma GG, Okada Y, Von Hoff D, Goel A. Non-coding RNA biomarkers in pancreatic ductal adenocarcinoma. Semin Cancer Biol 2021; 75: 153–168

[147]

Al-Shaheri FN, Alhamdani MSS, Bauer AS, Giese N, Büchler MW, Hackert T, Hoheisel JD. Blood biomarkers for differential diagnosis and early detection of pancreatic cancer. Cancer Treat Rev 2021; 96: 102193

[148]

Luo G, Jin K, Deng S, Cheng H, Fan Z, Gong Y, Qian Y, Huang Q, Ni Q, Liu C, Yu X. Roles of CA19-9 in pancreatic cancer: biomarker, predictor and promoter. Biochim Biophys Acta Rev Cancer 2021; 1875(2): 188409

[149]

Deng Y, Sun Z, Wang L, Wang M, Yang J, Li G. Biosensor-based assay of exosome biomarker for early diagnosis of cancer. Front Med 2022; 16(2): 157–175

[150]

Berger AW, Schwerdel D, Reinacher-Schick A, Uhl W, Algül H, Friess H, Janssen KP, König A, Ghadimi M, Gallmeier E, Bartsch DK, Geissler M, Staib L, Tannapfel A, Kleger A, Beutel A, Schulte LA, Kornmann M, Ettrich TJ, Seufferlein T. A blood-based multi marker assay supports the differential diagnosis of early-stage pancreatic cancer. Theranostics 2019; 9(5): 1280–1287

[151]

Thibault B, Ramos-Delgado F, Pons-Tostivint E, Therville N, Cintas C, Arcucci S, Cassant-Sourdy S, Reyes-Castellanos G, Tosolini M, Villard AV, Cayron C, Baer R, Bertrand-Michel J, Pagan D, Ferreira Da Mota D, Yan H, Falcomatà C, Muscari F, Bournet B, Delord JP, Aksoy E, Carrier A, Cordelier P, Saur D, Basset C, Guillermet-Guibert J. Pancreatic cancer intrinsic PI3Kα activity accelerates metastasis and rewires macrophage component. EMBO Mol Med 2021; 13(7): e13502

[152]

Groot VP, Mosier S, Javed AA, Teinor JA, Gemenetzis G, Ding D, Haley LM, Yu J, Burkhart RA, Hasanain A, Debeljak M, Kamiyama H, Narang A, Laheru DA, Zheng L, Lin MT, Gocke CD, Fishman EK, Hruban RH, Goggins MG, Molenaar IQ, Cameron JL, Weiss MJ, Velculescu VE, He J, Wolfgang CL, Eshleman JR. Circulating tumor DNA as a clinical test in resected pancreatic cancer. Clin Cancer Res 2019; 25(16): 4973–4984

[153]

Li H, Warden AR, Su W, He J, Zhi X, Wang K, Zhu L, Shen G, Ding X. Highly sensitive and portable mRNA detection platform for early cancer detection. J Nanobiotechnology 2021; 19(1): 287

[154]

Melo SA, Luecke LB, Kahlert C, Fernandez AF, Gammon ST, Kaye J, LeBleu VS, Mittendorf EA, Weitz J, Rahbari N, Reissfelder C, Pilarsky C, Fraga MF, Piwnica-Worms D, Kalluri R. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature 2015; 523(7559): 177–182

[155]

Nagata N, Nishijima S, Kojima Y, Hisada Y, Imbe K, Miyoshi-Akiyama T, Suda W, Kimura M, Aoki R, Sekine K, Ohsugi M, Miki K, Osawa T, Ueki K, Oka S, Mizokami M, Kartal E, Schmidt TSB, Molina-Montes E, Estudillo L, Malats N, Trebicka J, Kersting S, Langheinrich M, Bork P, Uemura N, Itoi T, Kawai T. Metagenomic identification of microbial signatures predicting pancreatic cancer from a multinational study. Gastroenterology 2022; 163(1): 222–238

[156]

Pang Y, Wang C, Lu L, Wang C, Sun Z, Xiao R. Dual-SERS biosensor for one-step detection of microRNAs in exosome and residual plasma of blood samples for diagnosing pancreatic cancer. Biosens Bioelectron 2019; 130: 204–213

[157]

Sharma GG, Okada Y, Von Hoff D, Goel A. Non-coding RNA biomarkers in pancreatic ductal adenocarcinoma. Semin Cancer Biol 2021; 75: 153–168

[158]

Jin F, Yang L, Wang W, Yuan N, Zhan S, Yang P, Chen X, Ma T, Wang Y. A novel class of tsRNA signatures as biomarkers for diagnosis and prognosis of pancreatic cancer. Mol Cancer 2021; 20(1): 95

[159]

Zhan S, Yang P, Zhou S, Xu Y, Xu R, Liang G, Zhang C, Chen X, Yang L, Jin F, Wang Y. Serum mitochondrial tsRNA serves as a novel biomarker for hepatocarcinoma diagnosis. Front Med 2022; 16(2): 216–226

[160]

Majumder S, Taylor WR, Foote PH, Berger CK, Wu CW, Mahoney DW, Bamlet WR, Burger KN, Postier N, de la Fuente J, Doering KA, Lidgard GP, Allawi HT, Petersen GM, Chari ST, Ahlquist DA, Kisiel JB. High detection rates of pancreatic cancer across stages by plasma assay of novel methylated DNA markers and CA19-9. Clin Cancer Res 2021; 27(9): 2523–2532

[161]

Kim Y, Yeo I, Huh I, Kim J, Han D, Jang JY, Kim Y. Development and multiple validation of the protein multi-marker panel for diagnosis of pancreatic cancer. Clin Cancer Res 2021; 27(8): 2236–2245

[162]

Mahajan UM, Oehrle B, Sirtl S, Alnatsha A, Goni E, Regel I, Beyer G, Vornhülz M, Vielhauer J, Chromik A, Bahra M, Klein F, Uhl W, Fahlbusch T, Distler M, Weitz J, Grützmann R, Pilarsky C, Weiss FU, Adam MG, Neoptolemos JP, Kalthoff H, Rad R, Christiansen N, Bethan B, Kamlage B, Lerch MM, Mayerle J. Independent validation and assay standardization of improved metabolic biomarker signature to differentiate pancreatic ductal adenocarcinoma from chronic pancreatitis. Gastroenterology 2022; 163(5): 1407–1422

[163]

Nam H, Hong SS, Jung KH, Kang S, Park MS, Kang S, Kim HS, Mai VH, Kim J, Lee H, Lee W, Suh YJ, Lim JH, Kim SY, Kim SC, Kim SH, Park S. A serum marker for early pancreatic cancer with a possible link to diabetes. J Natl Cancer Inst 2022; 114(2): 228–234

[164]

Wolrab D, Jirásko R, Cífková E, Höring M, Mei D, Chocholoušková M, Peterka O, Idkowiak J, Hrnčiarová T, Kuchař L, Ahrends R, Brumarová R, Friedecký D, Vivo-Truyols G, Škrha P, Škrha J, Kučera R, Melichar B, Liebisch G, Burkhardt R, Wenk MR, Cazenave-Gassiot A, Karásek P, Novotný I, Greplová K, Hrstka R, Holčapek M. Lipidomic profiling of human serum enables detection of pancreatic cancer. Nat Commun 2022; 13(1): 124

[165]

Staal B, Liu Y, Barnett D, Hsueh P, He Z, Gao C, Partyka K, Hurd MW, Singhi AD, Drake RR, Huang Y, Maitra A, Brand RE, Haab BB. The sTRA plasma biomarker: blinded validation of improved accuracy over CA19-9 in pancreatic cancer diagnosis. Clin Cancer Res 2019; 25(9): 2745–2754

[166]

Debernardi S, O’Brien H, Algahmdi AS, Malats N, Stewart GD, Plješa-Ercegovac M, Costello E, Greenhalf W, Saad A, Roberts R, Ney A, Pereira SP, Kocher HM, Duffy S, Blyuss O, Crnogorac-Jurcevic T. A combination of urinary biomarker panel and PancRISK score for earlier detection of pancreatic cancer: a case-control study. PLoS Med 2020; 17(12): e1003489

[167]

Nesteruk K, Levink IJM, de Vries E, Visser IJ, Peppelenbosch MP, Cahen DL, Fuhler GM, Bruno MJ. Extracellular vesicle-derived microRNAs in pancreatic juice as biomarkers for detection of pancreatic ductal adenocarcinoma. Pancreatology 2022; 22(5): 626–635

[168]

Kartal E, Schmidt TSB, Molina-Montes E, Rodríguez-Perales S, Wirbel J, Maistrenko OM, Akanni WA, Alashkar Alhamwe B, Alves RJ, Carrato A, Erasmus HP, Estudillo L, Finkelmeier F, Fullam A, Glazek AM, Gómez-Rubio P, Hercog R, Jung F, Kandels S, Kersting S, Langheinrich M, Márquez M, Molero X, Orakov A, Van Rossum T, Torres-Ruiz R, Telzerow A, Zych K; MAGIC Study investigators; PanGenEU Study investigators; Benes V, Zeller G, Trebicka J, Real FX, Malats N, Bork P. A faecal microbiota signature with high specificity for pancreatic cancer. Gut 2022; 71(7): 1359–1372

[169]

Kelly KN, Macedo FI, Merchant NB. Neoadjuvant therapy. Adv Surg 2020; 54: 49–68

[170]

Du Y, Ma Y, Zhu Q, Fu Y, Li Y, Zhang Y, Li M, Feng F, Yuan P, Wang X. GDF15 negatively regulates chemosensitivity via TGFBR2-AKT pathway-dependent metabolism in esophageal squamous cell carcinoma. Front Med 2023; 17(1): 119–131

[171]

Capula M, Perán M, Xu G, Donati V, Yee D, Gregori A, Assaraf YG, Giovannetti E, Deng D. Role of drug catabolism, modulation of oncogenic signaling and tumor microenvironment in microbe-mediated pancreatic cancer chemoresistance. Drug Resist Updat 2022; 64: 100864

[172]

Erkan M, Kleeff J, Gorbachevski A, Reiser C, Mitkus T, Esposito I, Giese T, Büchler MW, Giese NA, Friess H. Periostin creates a tumor-supportive microenvironment in the pancreas by sustaining fibrogenic stellate cell activity. Gastroenterology 2007; 132(4): 1447–1464

[173]

Yu M, Tannock IF. Targeting tumor architecture to favor drug penetration: a new weapon to combat chemoresistance in pancreatic cancer?. Cancer Cell 2012; 21(3): 327–329

[174]

Chamma H, Vila IK, Taffoni C, Turtoi A, Laguette N. Activation of STING in the pancreatic tumor microenvironment: a novel therapeutic opportunity. Cancer Lett 2022; 538: 215694

[175]

Huang Y, Kanada M, Ye J, Deng Y, He Q, Lei Z, Chen Y, Li Y, Qin P, Zhang J, Wei J. Exosome-mediated remodeling of the tumor microenvironment: from local to distant intercellular communication. Cancer Lett 2022; 543: 215796

[176]

Starling N, Hawkes EA, Chau I, Watkins D, Thomas J, Webb J, Brown G, Thomas K, Barbachano Y, Oates J, Cunningham D. A dose escalation study of gemcitabine plus oxaliplatin in combination with imatinib for gemcitabine-refractory advanced pancreatic adenocarcinoma. Ann Oncol 2012; 23(4): 942–947

[177]

Hu C, Xia R, Zhang X, Li T, Ye Y, Li G, He R, Li Z, Lin Q, Zheng S, Chen R. circFARP1 enables cancer-associated fibroblasts to promote gemcitabine resistance in pancreatic cancer via the LIF/STAT3 axis. Mol Cancer 2022; 21(1): 24

[178]

Malik S, Westcott JM, Brekken RA, Burrows FJ. CXCL12 in pancreatic cancer: its function and potential as a therapeutic drug target. Cancers (Basel) 2021; 14(1): 86

[179]

Wei L, Lin Q, Lu Y, Li G, Huang L, Fu Z, Chen R, Zhou Q. Cancer-associated fibroblasts-mediated ATF4 expression promotes malignancy and gemcitabine resistance in pancreatic cancer via the TGF-β1/SMAD2/3 pathway and ABCC1 transactivation. Cell Death Dis 2021; 12(4): 334

[180]

Zhang X, Zheng S, Hu C, Li G, Lin H, Xia R, Ye Y, He R, Li Z, Lin Q, Chen R, Zhou Q. Cancer-associated fibroblast-induced lncRNA UPK1A-AS1 confers platinum resistance in pancreatic cancer via efficient double-strand break repair. Oncogene 2022; 41(16): 2372–2389

[181]

Guo Y, Wu H, Xiong J, Gou S, Cui J, Peng T. miR-222-3p-containing macrophage-derived extracellular vesicles confer gemcitabine resistance via TSC1-mediated mTOR/AKT/PI3K pathway in pancreatic cancer. Cell Biol Toxicol 2023; 39(4): 1203–1214

[182]

Halbrook CJ, Pontious C, Kovalenko I, Lapienyte L, Dreyer S, Lee HJ, Thurston G, Zhang Y, Lazarus J, Sajjakulnukit P, Hong HS, Kremer DM, Nelson BS, Kemp S, Zhang L, Chang D, Biankin A, Shi J, Frankel TL, Crawford HC, Morton JP, Pasca di Magliano M, Lyssiotis CA. Macrophage-released pyrimidines inhibit gemcitabine therapy in pancreatic cancer. Cell Metab 2019; 29(6): 1390–1399.e6

[183]

Huanwen W, Zhiyong L, Xiaohua S, Xinyu R, Kai W, Tonghua L. Intrinsic chemoresistance to gemcitabine is associated with constitutive and laminin-induced phosphorylation of FAK in pancreatic cancer cell lines. Mol Cancer 2009; 8: 125

[184]

Richards KE, Zeleniak AE, Fishel ML, Wu J, Littlepage LE, Hill R. Cancer-associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Oncogene 2017; 36(13): 1770–1778

[185]

Richards KE, Xiao W, Hill R, On Behalf Of The Usc Pancreas Research Team. Cancer-associated fibroblasts confer gemcitabine resistance to pancreatic cancer cells through PTEN-targeting miRNAs in exosomes. Cancers (Basel) 2022; 14(11): 2812

[186]

Patel GK, Khan MA, Bhardwaj A, Srivastava SK, Zubair H, Patton MC, Singh S, Khushman M, Singh AP. Exosomes confer chemoresistance to pancreatic cancer cells by promoting ROS detoxification and miR-155-mediated suppression of key gemcitabine-metabolising enzyme, DCK. Br J Cancer 2017; 116(5): 609–619

[187]

Chan MK, Chung JY, Tang PC, Chan AS, Ho JY, Lin TP, Chen J, Leung KT, To KF, Lan HY, Tang PM. TGF-β signaling networks in the tumor microenvironment. Cancer Lett 2022; 550: 215925

[188]

Cioffi M, Trabulo SM, Sanchez-Ripoll Y, Miranda-Lorenzo I, Lonardo E, Dorado J, Reis Vieira C, Ramirez JC, Hidalgo M, Aicher A, Hahn S, Sainz B Jr, Heeschen C. The miR-17-92 cluster counteracts quiescence and chemoresistance in a distinct subpopulation of pancreatic cancer stem cells. Gut 2015; 64(12): 1936–1948

[189]

Yang MC, Wang HC, Hou YC, Tung HL, Chiu TJ, Shan YS. Blockade of autophagy reduces pancreatic cancer stem cell activity and potentiates the tumoricidal effect of gemcitabine. Mol Cancer 2015; 14(1): 179

[190]

Zheng X, Carstens JL, Kim J, Scheible M, Kaye J, Sugimoto H, Wu CC, LeBleu VS, Kalluri R. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature 2015; 527(7579): 525–530

[191]

Tang B, Yang Y, Kang M, Wang Y, Wang Y, Bi Y, He S, Shimamoto F. m6A demethylase ALKBH5 inhibits pancreatic cancer tumorigenesis by decreasing WIF-1 RNA methylation and mediating Wnt signaling. Mol Cancer 2020; 19(1): 3

[192]

Akada M, Crnogorac-Jurcevic T, Lattimore S, Mahon P, Lopes R, Sunamura M, Matsuno S, Lemoine NR. Intrinsic chemoresistance to gemcitabine is associated with decreased expression of BNIP3 in pancreatic cancer. Clin Cancer Res 2005; 11(8): 3094–3101

[193]

Gu J, Huang W, Wang X, Zhang J, Tao T, Zheng Y, Liu S, Yang J, Chen ZS, Cai CY, Li J, Wang H, Fan Y. Hsa-miR-3178/RhoB/PI3K/Akt, a novel signaling pathway regulates ABC transporters to reverse gemcitabine resistance in pancreatic cancer. Mol Cancer 2022; 21(1): 112

[194]

Zhou C, Yi C, Yi Y, Qin W, Yan Y, Dong X, Zhang X, Huang Y, Zhang R, Wei J, Ali DW, Michalak M, Chen XZ, Tang J. LncRNA PVT1 promotes gemcitabine resistance of pancreatic cancer via activating Wnt/β-catenin and autophagy pathway through modulating the miR-619-5p/Pygo2 and miR-619-5p/ATG14 axes. Mol Cancer 2020; 19: 118

[195]

Xiong G, Liu C, Yang G, Feng M, Xu J, Zhao F, You L, Zhou L, Zheng L, Hu Y, Wang X, Zhang T, Zhao Y. Long noncoding RNA GSTM3TV2 upregulates LAT2 and OLR1 by competitively sponging let-7 to promote gemcitabine resistance in pancreatic cancer. J Hematol Oncol 2019; 12(1): 97

[196]

Chen ZW, Hu JF, Wang ZW, Liao CY, Kang FP, Lin CF, Huang Y, Huang L, Tian YF, Chen S. Circular RNA circ-MTHFD1L induces HR repair to promote gemcitabine resistance via the miR-615-3p/RPN6 axis in pancreatic ductal adenocarcinoma. J Exp Clin Cancer Res 2022; 41(1): 153

[197]

Tu M, Li H, Lv N, Xi C, Lu Z, Wei J, Chen J, Guo F, Jiang K, Song G, Gao W, Miao Y. Vasohibin 2 reduces chemosensitivity to gemcitabine in pancreatic cancer cells via Jun proto-oncogene dependent transactivation of ribonucleotide reductase regulatory subunit M2. Mol Cancer 2017; 16(1): 66

[198]

Biliran H Jr, Wang Y, Banerjee S, Xu H, Heng H, Thakur A, Bollig A, Sarkar FH, Liao JD. Overexpression of cyclin D1 promotes tumor cell growth and confers resistance to cisplatin-mediated apoptosis in an elastase-myc transgene-expressing pancreatic tumor cell line. Clin Cancer Res 2005; 11(16): 6075–6086

[199]

Melisi D, Xia Q, Paradiso G, Ling J, Moccia T, Carbone C, Budillon A, Abbruzzese JL, Chiao PJ. Modulation of pancreatic cancer chemoresistance by inhibition of TAK1. J Natl Cancer Inst 2011; 103(15): 1190–1204

[200]

Kadera BE, Toste PA, Wu N, Li L, Nguyen AH, Dawson DW, Donahue TR. Low expression of the E3 ubiquitin ligase CBL confers chemoresistance in human pancreatic cancer and is targeted by epidermal growth factor receptor inhibition. Clin Cancer Res 2015; 21(1): 157–165

[201]

Wu L, Ge Y, Yuan Y, Li H, Sun H, Xu C, Wang Y, Zhao T, Wang X, Liu J, Gao S, Chang A, Hao J, Huang C. Genome-wide CRISPR screen identifies MTA3 as an inducer of gemcitabine resistance in pancreatic ductal adenocarcinoma. Cancer Lett 2022; 548: 215864

[202]

Safarzadeh Kozani P, Safarzadeh Kozani P, Rahbarizadeh F. CAR T cells redirected against tumor-specific antigen glycoforms: can low-sugar antigens guarantee a sweet success?. Front Med 2022; 16(3): 322–338

[203]

Safarzadeh Kozani P, Safarzadeh Kozani P, Rahbarizadeh F. CAR T cells redirected against tumor-specific antigen glycoforms: can low-sugar antigens guarantee a sweet success?. Front Med 2022; 16(3): 322–338

[204]

Yang K, Li J, Zhao L, Sun Z, Bai C. Estimating the number of Chinese cancer patients eligible for and benefit from immune checkpoint inhibitors. Front Med 2022; 16(5): 773–783

[205]

Xu R, Du S, Zhu J, Meng F, Liu B. Neoantigen-targeted TCR-T cell therapy for solid tumors: how far from clinical application. Cancer Lett 2022; 546: 215840

[206]

Bockorny B, Grossman JE, Hidalgo M. Facts and hopes in immunotherapy of pancreatic cancer. Clin Cancer Res 2022; 28(21): 4606–4617

[207]

Zhu YH, Zheng JH, Jia QY, Duan ZH, Yao HF, Yang J, Sun YW, Jiang SH, Liu DJ, Huo YM. Immunosuppression, immune escape, and immunotherapy in pancreatic cancer: focused on the tumor microenvironment. Cell Oncol (Dordr) 2023; 46(1): 17–48

[208]

Ostios-Garcia L, Villamayor J, Garcia-Lorenzo E, Vinal D, Feliu J. Understanding the immune response and the current landscape of immunotherapy in pancreatic cancer. World J Gastroenterol 2021; 27(40): 6775–6793

[209]

Chen TW, Hung WZ, Chiang SF, Chen WT, Ke TW, Liang JA, Huang CY, Yang PC, Huang KC, Chao KSC. Dual inhibition of TGFβ signaling and CSF1/CSF1R reprograms tumor-infiltrating macrophages and improves response to chemotherapy via suppressing PD-L1. Cancer Lett 2022; 543: 215795

[210]

Hadlandsmyth K, Conrad M, Steffensmeier KS, Van Tiem J, Obrecht A, Cullen JJ, Vander Weg MW. Enhancing the biopsychosocial approach to perioperative care: a pilot randomized trial of the perioperative pain self-management (PePS) intervention. Ann Surg 2022; 275(1): e8–e14

[211]

Wang H, Shao Q, Wang J, Zhao L, Wang L, Cheng Z, Yue C, Chen W, Wang H, Zhang Y. Decreased CXCR2 expression on circulating monocytes of colorectal cancer impairs recruitment and induces Re-education of tumor-associated macrophages. Cancer Lett 2022; 529: 112–125

[212]

Chen QY, Gao B, Tong D, Huang C. Crosstalk between extracellular vesicles and tumor-associated macrophage in the tumor microenvironment. Cancer Lett 2023; 552: 215979

[213]

Pylayeva-Gupta Y, Lee KE, Hajdu CH, Miller G, Bar-Sagi D. Oncogenic Kras-induced GM-CSF production promotes the development of pancreatic neoplasia. Cancer Cell 2012; 21(6): 836–847

[214]

Monti P, Leone BE, Marchesi F, Balzano G, Zerbi A, Scaltrini F, Pasquali C, Calori G, Pessi F, Sperti C, Di Carlo V, Allavena P, Piemonti L. The CC chemokine MCP-1/CCL2 in pancreatic cancer progression: regulation of expression and potential mechanisms of antimalignant activity. Cancer Res 2003; 63(21): 7451–7461

[215]

Li J, Byrne KT, Yan F, Yamazoe T, Chen Z, Baslan T, Richman LP, Lin JH, Sun YH, Rech AJ, Balli D, Hay CA, Sela Y, Merrell AJ, Liudahl SM, Gordon N, Norgard RJ, Yuan S, Yu S, Chao T, Ye S, Eisinger-Mathason TSK, Faryabi RB, Tobias JW, Lowe SW, Coussens LM, Wherry EJ, Vonderheide RH, Stanger BZ. Tumor cell-intrinsic factors underlie heterogeneity of immune cell infiltration and response to immunotherapy. Immunity 2018; 49(1): 178–193.e7

[216]

Zhang A, Qian Y, Ye Z, Chen H, Xie H, Zhou L, Shen Y, Zheng S. Cancer-associated fibroblasts promote M2 polarization of macrophages in pancreatic ductal adenocarcinoma. Cancer Med 2017; 6(2): 463–470

[217]

Yang X, Lin Y, Shi Y, Li B, Liu W, Yin W, Dang Y, Chu Y, Fan J, He R. FAP promotes immunosuppression by cancer-associated fibroblasts in the tumor microenvironment via STAT3-CCL2 signaling. Cancer Res 2016; 76(14): 4124–4135

[218]

Yu Y. Multi-target combinatory strategy to overcome tumor immune escape. Front Med 2022; 16(2): 208–215

[219]

Traub B, Link KH, Kornmann M. Curing pancreatic cancer. Semin Cancer Biol 2021; 76: 232–246

[220]

Hackert T, Sachsenmaier M, Hinz U, Schneider L, Michalski CW, Springfeld C, Strobel O, Jäger D, Ulrich A, Büchler MW. Locally advanced pancreatic cancer: neoadjuvant therapy with folfirinox results in resectability in 60% of the patients. Ann Surg 2016; 264(3): 457–463

[221]

Murphy JE, Wo JY, Ryan DP, Clark JW, Jiang W, Yeap BY, Drapek LC, Ly L, Baglini CV, Blaszkowsky LS, Ferrone CR, Parikh AR, Weekes CD, Nipp RD, Kwak EL, Allen JN, Corcoran RB, Ting DT, Faris JE, Zhu AX, Goyal L, Berger DL, Qadan M, Lillemoe KD, Talele N, Jain RK, DeLaney TF, Duda DG, Boucher Y, Fernández-Del Castillo C, Hong TS. Total neoadjuvant therapy with FOLFIRINOX in combination with losartan followed by chemoradiotherapy for locally advanced pancreatic cancer: a phase 2 clinical trial. JAMA Oncol 2019; 5(7): 1020–1027

[222]

Murphy JE, Wo JY, Ryan DP, Jiang W, Yeap BY, Drapek LC, Blaszkowsky LS, Kwak EL, Allen JN, Clark JW, Faris JE, Zhu AX, Goyal L, Lillemoe KD, DeLaney TF, Fernández-Del Castillo C, Ferrone CR, Hong TS. Total neoadjuvant therapy with FOLFIRINOX followed by individualized chemoradiotherapy for borderline resectable pancreatic adenocarcinoma: a phase 2 clinical trial. JAMA Oncol 2018; 4(7): 963–969

[223]

Hackert T, Niesen W, Hinz U, Tjaden C, Strobel O, Ulrich A, Michalski CW, Büchler MW. Radical surgery of oligometastatic pancreatic cancer. Eur J Surg Oncol 2017; 43(2): 358–363

[224]

Tachezy M, Gebauer F, Janot M, Uhl W, Zerbi A, Montorsi M, Perinel J, Adham M, Dervenis C, Agalianos C, Malleo G, Maggino L, Stein A, Izbicki JR, Bockhorn M. Synchronous resections of hepatic oligometastatic pancreatic cancer: disputing a principle in a time of safe pancreatic operations in a retrospective multicenter analysis. Surgery 2016; 160(1): 136–144

[225]

Strobel O, Neoptolemos J, Jäger D, Büchler MW. Optimizing the outcomes of pancreatic cancer surgery. Nat Rev Clin Oncol 2019; 16(1): 11–26

[226]

Valle JW, Palmer D, Jackson R, Cox T, Neoptolemos JP, Ghaneh P, Rawcliffe CL, Bassi C, Stocken DD, Cunningham D, O’Reilly D, Goldstein D, Robinson BA, Karapetis C, Scarfe A, Lacaine F, Sand J, Izbicki JR, Mayerle J, Dervenis C, Oláh A, Butturini G, Lind PA, Middleton MR, Anthoney A, Sumpter K, Carter R, Büchler MW. Optimal duration and timing of adjuvant chemotherapy after definitive surgery for ductal adenocarcinoma of the pancreas: ongoing lessons from the ESPAC-3 study. J Clin Oncol 2014; 32(6): 504–512

[227]

Nassour I, Wang SC, Christie A, Augustine MM, Porembka MR, Yopp AC, Choti MA, Mansour JC, Xie XJ, Polanco PM, Minter RM. Minimally invasive versus open pancreaticoduodenectomy: a propensity-matched study from a national cohort of patients. Ann Surg 2018; 268(1): 151–157

[228]

Raoof M, Ituarte PHG, Woo Y, Warner SG, Singh G, Fong Y, Melstrom L. Propensity score-matched comparison of oncological outcomes between laparoscopic and open distal pancreatic resection. Br J Surg 2018; 105(5): 578–586

[229]

van Hilst J, de Rooij T, Klompmaker S, Rawashdeh M, Aleotti F, Al-Sarireh B, Alseidi A, Ateeb Z, Balzano G, Berrevoet F, Björnsson B, Boggi U, Busch OR, Butturini G, Casadei R, Del Chiaro M, Chikhladze S, Cipriani F, van Dam R, Damoli I, van Dieren S, Dokmak S, Edwin B, van Eijck C, Fabre JM, Falconi M, Farges O, Fernández-Cruz L, Forgione A, Frigerio I, Fuks D, Gavazzi F, Gayet B, Giardino A, Groot Koerkamp B, Hackert T, Hassenpflug M, Kabir I, Keck T, Khatkov I, Kusar M, Lombardo C, Marchegiani G, Marshall R, Menon KV, Montorsi M, Orville M, de Pastena M, Pietrabissa A, Poves I, Primrose J, Pugliese R, Ricci C, Roberts K, Røsok B, Sahakyan MA, Sánchez-Cabús S, Sandström P, Scovel L, Solaini L, Soonawalla Z, Souche FR, Sutcliffe RP, Tiberio GA, Tomazic A, Troisi R, Wellner U, White S, Wittel UA, Zerbi A, Bassi C, Besselink MG, Abu Hilal M; European Consortium on Minimally Invasive Pancreatic Surgery (E-MIPS). Minimally invasive versus open distal pancreatectomy for ductal adenocarcinoma (DIPLOMA): a pan-European propensity score matched study. Ann Surg 2019; 269(1): 10–17

[230]

Tempero MA, Malafa MP, Al-Hawary M, Behrman SW, Benson AB, Cardin DB, Chiorean EG, Chung V, Czito B, Del Chiaro M, Dillhoff M, Donahue TR, Dotan E, Ferrone CR, Fountzilas C, Hardacre J, Hawkins WG, Klute K, Ko AH, Kunstman JW, LoConte N, Lowy AM, Moravek C, Nakakura EK, Narang AK, Obando J, Polanco PM, Reddy S, Reyngold M, Scaife C, Shen J, Vollmer C, Wolff RA, Wolpin BM, Lynn B, George GV. Pancreatic Adenocarcinoma, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2021; 19(4): 439–457

[231]

Zhang Y, Yang C, Cheng H, Fan Z, Huang Q, Lu Y, Fan K, Luo G, Jin K, Wang Z, Liu C, Yu X. Novel agents for pancreatic ductal adenocarcinoma: emerging therapeutics and future directions. J Hematol Oncol 2018; 11(1): 14

[232]

Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Kamiyama H, Jimeno A, Hong SM, Fu B, Lin MT, Calhoun ES, Kamiyama M, Walter K, Nikolskaya T, Nikolsky Y, Hartigan J, Smith DR, Hidalgo M, Leach SD, Klein AP, Jaffee EM, Goggins M, Maitra A, Iacobuzio-Donahue C, Eshleman JR, Kern SE, Hruban RH, Karchin R, Papadopoulos N, Parmigiani G, Vogelstein B, Velculescu VE, Kinzler KW. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 2008; 321(5897): 1801–1806

[233]

Zhong J, Bai H, Wang Z, Duan J, Zhuang W, Wang D, Wan R, Xu J, Fei K, Ma Z, Zhang X, Wang J. Treatment of advanced non-small cell lung cancer with driver mutations: current applications and future directions. Front Med 2023; 17(1): 18–42

[234]

Zhao H, Luo F, Xue J, Li S, Xu RH. Emerging immunological strategies: recent advances and future directions. Front Med 2021; 15(6): 805–828

[235]

Mi JQ, Xu J, Zhou J, Zhao W, Chen Z, Melenhorst JJ, Chen S. CAR T-cell immunotherapy: a powerful weapon for fighting hematological B-cell malignancies. Front Med 2021; 15(6): 783–804

[236]

Chen S, Zhao W, Li J, Wu D; Lymphoid Disease Group, Chinese Society of Hematology, Chinese Medical Association. Chinese expert consensus on oral drugs for the treatment of mature B-cell lymphomas (2020 edition). Front Med 2022; 16(5): 815–826

[237]

Bear AS, Vonderheide RH, O’Hara MH. Challenges and opportunities for pancreatic cancer immunotherapy. Cancer Cell 2020; 38(6): 788–802

[238]

Li Y, Wang S, Lin M, Hou C, Li C, Li G. Analysis of interactions of immune checkpoint inhibitors with antibiotics in cancer therapy. Front Med 2022; 16(3): 307–321

[239]

Heumann T, Judkins C, Li K, Lim SJ, Hoare J, Parkinson R, Cao H, Zhang T, Gai J, Celiker B, Zhu Q, McPhaul T, Durham J, Purtell K, Klein R, Laheru D, De Jesus-Acosta A, Le DT, Narang A, Anders R, Burkhart R, Burns W, Soares K, Wolfgang C, Thompson E, Jaffee E, Wang H, He J, Zheng L. A platform trial of neoadjuvant and adjuvant antitumor vaccination alone or in combination with PD-1 antagonist and CD137 agonist antibodies in patients with resectable pancreatic adenocarcinoma. Nat Commun 2023; 14(1): 3650

RIGHTS & PERMISSIONS

Higher Education Press

AI Summary AI Mindmap
PDF (3106KB)

3201

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/