1
|
Zhang P, Pan J, Lin S, Peng B, An C, Zhang J, Xu L, Lai Y, Yu H, Xu Z. Smart drug delivery platforms reprogramming cancer immune cycle to mitigate immune resistance of pancreatic tumors. Adv Drug Deliv Rev 2025; 224:115620. [DOI: 10.1016/j.addr.2025.115620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2025]
|
2
|
Liu H, Zhao Z, Wu C, Chen J, He Z, Jiang K. ZMYND8 promotes the Warburg effect and tumorigenesis through c-Myc activation in pancreatic cancer. Oncogene 2025:10.1038/s41388-025-03483-0. [PMID: 40579459 DOI: 10.1038/s41388-025-03483-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 05/27/2025] [Accepted: 06/18/2025] [Indexed: 06/29/2025]
Abstract
Pancreatic cancer (PC) is a digestive tract tumour with an extremely poor patient prognosis and prominent metabolic abnormalities. However, the molecular mechanisms underlying metabolic reprogramming in the progression of pancreatic cancer remain poorly understood. Here, we employed an epigenetic siRNA library to identify a crucial regulator, ZMYND8, which is involved in glycolysis in PC cells. ZMYND8 was frequently overexpressed in both PC tissues and cell lines, and its elevated expression was significantly correlated with poor overall survival in patients with PC. The high rates of glucose uptake and lactate secretion conferred by ZMYND8 revealed an abnormal activity of aerobic glycolysis in PC cells. Functional studies revealed that ZMYND8 significantly promoted the proliferation, migration and invasion of PC cells. Integrated analyses of CUT&Tag and RNA-seq data revealed that ZMYND8 may activate c-Myc transcriptional activity by modulating downstream epigenetic regulatory pathways. Proteomic profiling and coimmunoprecipitation (Co-IP) assays further demonstrated a direct physical interaction between ZMYND8 and c-Myc. Mechanistic studies revealed that ZMYND8 interacted with and activated c-Myc, thereby promoting the Warburg effect and facilitating PC cell malignancy. Moreover, in vivo studies revealed that overexpression of ZMYND8 resulted in accelerated tumour growth in PC xenografts, which was reversible through the knockdown of c-Myc or treatment with 2-deoxy-D-glucose. Collectively, our data suggest that ZMYND8 functions as a critical metabolic regulator in PC cells by tightly regulating c-Myc activity and may represent a promising novel therapeutic target for advanced pancreatic cancer treatment.
Collapse
Affiliation(s)
- Hui Liu
- Medical School of Chinese PLA, Beijing, PR China
- Faculty of Hepatopancreatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, PR China
- Department of General Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, PR China
| | - Zhifeng Zhao
- Medical School of Chinese PLA, Beijing, PR China
- Faculty of Hepatopancreatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, PR China
| | - Changle Wu
- Medical School of Chinese PLA, Beijing, PR China
- Faculty of Hepatopancreatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, PR China
| | - Jinxin Chen
- Department of Hepatobiliary Surgery, Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy Center, Shenzhen University, Shenzhen, Guangdong, PR China
| | - Zhiwei He
- Department of Hepatobiliary Surgery, Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy Center, Shenzhen University, Shenzhen, Guangdong, PR China.
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, PR China.
| | - Kai Jiang
- Medical School of Chinese PLA, Beijing, PR China.
- Faculty of Hepatopancreatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, PR China.
| |
Collapse
|
3
|
Li M, Yu X, Liu Y, Ouyang S, Wu L, Chen X, Yu H, Chen H, Lian S, Li Z, Gong L, Song L, Li J. KRAS/ABHD17C/ALOX15B Axis Promotes Pancreatic Cancer Progression via Ferroptosis Evasion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e04470. [PMID: 40569151 DOI: 10.1002/advs.202504470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 06/09/2025] [Indexed: 06/28/2025]
Abstract
Understanding the mechanisms underlying Kirsten rat sarcoma (KRAS) mutation-driven development and progression of pancreatic ductal adenocarcinoma (PDAC) may facilitate the discovery of novel strategies for KRAS-mutant PDAC (KRASmut-PDAC) treatment. Here, it is reported that downregulation of arachidonate 15-lipoxygenase (ALOX15B) significantly correlated with poor outcomes in patients with KRASmut-PDAC. Mechanistically, KRASmut/ERK1-elicited phosphorylation of ABHD17C promotes depalmitoylation and membrane-to-cytoplasm translocation of ALOX15B, facilitating proteasome-dependent degradation of ALOX15B via interaction with the E3 ligase complex CUL4/DDB1/DCAF10. Notably, treatment with methyl protodioscin (MPD), a steroid saponin primarily purified from polygonatum sibiricum rhizome, restored the S-palmitoylation and membrane location of ALOX15B via disruption of the ABHD17C/ALOX15B interaction, consequently resulting in significant inhibition of growth rate of patient-derived KRASmut-PDAC organoids in vitro and KRASmut-PDAC-formed tumor in vivo via induction of ferroptosis. Therefore, these findings unveil a prominent role of ferroptosis evasion in KRASmut-PDAC progression and highlight the potential of targeting KRAS/ERK1/ABHD17C/ALOX15B axis in KRASmut-PDAC treatment.
Collapse
Affiliation(s)
- Man Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510080, China
- Department of Biochemistry, Zhongshan School of Medicine Sun Yat-sen University, Guangzhou, 510080, China
| | - Xuexin Yu
- Department of Biochemistry, Zhongshan School of Medicine Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuanji Liu
- Department of Biochemistry, Zhongshan School of Medicine Sun Yat-sen University, Guangzhou, 510080, China
| | - Shuqin Ouyang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510080, China
| | - Long Wu
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiaohong Chen
- Department of Biochemistry, Zhongshan School of Medicine Sun Yat-sen University, Guangzhou, 510080, China
| | - Huiqi Yu
- Department of Biochemistry, Zhongshan School of Medicine Sun Yat-sen University, Guangzhou, 510080, China
| | - Haoming Chen
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Senmao Lian
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ziwen Li
- Department of Biochemistry, Zhongshan School of Medicine Sun Yat-sen University, Guangzhou, 510080, China
| | - Liyun Gong
- Guangdong Provincial Key Laboratory for Genome Stability and Disease Prevention, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518061, China
| | - Libing Song
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510080, China
| | - Jun Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510080, China
- Department of Biochemistry, Zhongshan School of Medicine Sun Yat-sen University, Guangzhou, 510080, China
| |
Collapse
|
4
|
Duttenhefner JN, Reindl KM. GSTP1 knockdown induces metabolic changes affecting energy production and lipid balance in pancreatic cancer cells. Mol Cell Oncol 2025; 12:2518773. [PMID: 40524738 PMCID: PMC12169041 DOI: 10.1080/23723556.2025.2518773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2025] [Revised: 06/06/2025] [Accepted: 06/09/2025] [Indexed: 06/19/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with limited treatment options, underscoring the need for novel therapeutic targets. Metabolic reprogramming is a hallmark of PDAC, enabling tumor cells to sustain rapid proliferation and survive under nutrient-deprived conditions. While glutathione S-transferase pi 1 (GSTP1) is a known regulator of redox homeostasis in PDAC, its role in metabolic adaptation remains unclear. Here, we show that GSTP1 knockdown disrupts PDAC metabolism, leading to downregulation of key metabolic enzymes (ALDH7A1, CPT1A, SLC2A3, PGM1), ATP depletion, mitochondrial dysfunction, and phospholipid remodeling. Phospholipid remodeling, including an increase in phosphatidylcholine (PC) levels, further suggests a compensatory response to metabolic stress. Importantly, GSTP1 knockdown led to elevated lipid peroxidation, increasing 4-hydroxynonenal (4-HNE) accumulation. Treatment with the antioxidant N-acetyl cysteine (NAC) partially restored metabolic gene expression, reinforcing GSTP1's role in the interplay between redox regulation and metabolism in PDAC. By disrupting multiple metabolic pathways, GSTP1 depletion creates potential therapeutic vulnerabilities that could be targeted through metabolic and oxidative stress-inducing therapies to enhance treatment efficacy.
Collapse
Affiliation(s)
| | - Katie M. Reindl
- Department of Biological Sciences, North Dakota State University, Fargo, ND, USA
| |
Collapse
|
5
|
Iovanna J, Estaras M, Grasso D, Fernández Zapico ME, Neira JL, Santofimia-Castaño P. Oncogenic stress response mechanisms as new therapeutic targets in cancer treatment: A review. Medicine (Baltimore) 2025; 104:e42857. [PMID: 40527836 DOI: 10.1097/md.0000000000042857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/19/2025] Open
Abstract
Activation of oncogenes, such as through mutations in Kirsten rat sarcoma viral oncogene homolog (KRAS), triggers profound disruptions in cellular homeostasis that set off a cascade of stress responses. These responses enable cells to cope with the array of challenges encountered during tumorigenesis by activating defense mechanisms that promote adaptation and survival. Key components of this oncogenic stress response include heat shock proteins, the ubiquitin-proteasome system, autophagy, nuclear factor erythroid 2-related factor 2-antioxidant response element signaling, DNA damage response proteins, p53, redox-regulating proteins, and stress granules. This review concentrates on KRAS-driven oncogenic transformation, as KRAS mutations are among the most common in human cancers, accounting for over 90% of pancreatic ductal adenocarcinoma cases, around 30% of lung cancers, and approximately 50% of colorectal cancers. We examine the intricate molecular interplay between oncogenic stress and the associated cellular defense mechanisms, emphasizing the key molecular events that follow KRAS activation. Importantly, the very pathways that allow cancer cells to adapt to oncogenic stress also offer novel therapeutic opportunities. By selectively targeting pivotal regulators within these stress response pathways, we can potentially disrupt the survival mechanisms of cancer cells. This strategy not only promises to enhance the effectiveness of existing treatments but also paves the way for the development of innovative therapies designed to combat tumor progression. In essence, exploiting oncogenic stress responses represents an original and promising therapeutic approach in the fight against cancer.
Collapse
Affiliation(s)
- Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
- Hospital de Alta Complejidad El Cruce, Florencio Varela, Argentina
- University Arturo Jauretche, Florencio Varela, Argentina
| | - Matías Estaras
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Daniel Grasso
- Facultad de Farmacia y Bioquímica, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | - Jose Luis Neira
- Instituto de Biocomputación y Física de Sistemas Complejos, Unidad mixta GBsC-CSIC-BIFI, Zaragoza, Spain
- IDIBE, Universidad Miguel Hernández, Edificio Torregaitán, Elche, Spain
| | - Patricia Santofimia-Castaño
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| |
Collapse
|
6
|
Dong P, Ni J, Zheng X, Wang M, Yang M, Han H. Small molecules for Kirsten rat sarcoma viral oncogene homolog mutant cancers: Past, present, and future. Eur J Pharmacol 2025; 996:177428. [PMID: 40024323 DOI: 10.1016/j.ejphar.2025.177428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations have been identified in more than 20% of human cancers as one of the most common oncogenes, especially in non-small cell lung, colorectal, and pancreatic cancers. KRAS regulates the activation of multiple proteins involved in cell growth and proliferation, such as extracellular regulated protein kinases and mammalian target of rapamycin, as a hub between the epidermal growth factor receptor (EGFR) and downstream MAPK and AKT pathways. However, due to the lack of a binding pocket, KRAS has long been considered an undruggable target in recent decades until the discovery of Sotorasib (AMG510). With the approval of Glecirasib (JAB-21822), there are three approved small molecule inhibitors of KRAS, all of which are KRAS G12C inhibitors. At the same time, the limited clinical benefits and rapid emergence of drug resistance to the approved inhibitors have also promoted the emergence of more therapeutics, such as tri-complexes and proteolysis-targeting chimeras (PROTAC). In this paper, we summarize the development of KRAS inhibitors (KRASG12C, KRASG12D, and KRASmulti inhibitors, PROTAC, and tri-complex) and discuss the challenges and opportunities in the discovery of KRAS inhibitors in the hope of providing insights into the development of novel medications for KRAS.
Collapse
Affiliation(s)
- Peiliang Dong
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Jiating Ni
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin, 150040, China
| | - Xinyue Zheng
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin, 150040, China
| | - Mingtao Wang
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin, 150040, China
| | - Meng Yang
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin, 150040, China
| | - Hua Han
- Key Laboratory of Chinese Materia Medica, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin, 150040, China.
| |
Collapse
|
7
|
Gupta T, Murtaza M. Advancing targeted therapies in pancreatic cancer: Leveraging molecular abberrations for therapeutic success. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 196:19-32. [PMID: 39988056 DOI: 10.1016/j.pbiomolbio.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 02/03/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Pancreatic cancer is one of the most deadly with poor prognosis and overall survival rate due to the dense stroma in the tumors which often is challenging for the delivery of drug to penetrate deep inside the tumor bed and usually results in the progression of cancer. The conventional treatment such as chemotherapy, radiotherapy or surgery shows a minimal benefit in the survival due to the drug resistance, poor penetration, less radiosensitivity or recurrence of tumor. There is an urgent demand to develop molecular-level targeted therapies to achieve therapeutic efficacy in the pancreatic ductal adenocarcinoma (PDAC) patients. The precision oncology focuses on the unique attributes of the patient such as epigenome, proteome, genome, microbiome, lifestyle and diet habits which contributes to promote oncogenesis. The targeted therapy helps to target the mutated proteins responsible for controlling growth, division and metastasis of tumor in the cancer cells. It is very important to consider all the attributes of the patient to provide the suitable personalized treatment to avoid any severe side effects. In this review, we have laid emphasis on the precision medicine; the utmost priority is to improve the survival of cancer patients by targeting molecular mutations through transmembrane proteins, inhibitors, signaling pathways, immunotherapy, gene therapy or the use of nanocarriers for the delivery at the tumor site. It will become beneficial therapeutic window to be considered for the advanced stage pancreatic cancer patients to prolong their survival rate.
Collapse
Affiliation(s)
- Tanvi Gupta
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 704, Taiwan.
| | - Mohd Murtaza
- Fermentation & Microbial Biotechnology Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180016, India.
| |
Collapse
|
8
|
Roichman A, Zuo Q, Hwang S, Lu W, Cordova RA, MacArthur MR, Boyer JA, Mitchell SJ, Powers J, Koval SA, Hunter CJ, Rijmers J, Ryseck RP, AbuSalim JE, Chatterjee S, Lee WD, Xu X, Xing X, Chen Z, Zeng X, Marwaha S, McBride MJ, Guo JY, Kang Y, Donia MS, Rabinowitz JD. Microbiome metabolism of dietary phytochemicals controls the anticancer activity of PI3K inhibitors. Cell 2025; 188:3065-3080.e21. [PMID: 40393457 DOI: 10.1016/j.cell.2025.04.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 03/04/2025] [Accepted: 04/29/2025] [Indexed: 05/22/2025]
Abstract
Phosphatidylinositol 3-kinase (PI3K) signaling is both the effector pathway of insulin and among the most frequently activated pathways in human cancer. In murine cancer models, the efficacy of PI3K inhibitors is dramatically enhanced by a ketogenic diet, with a proposed mechanism involving dietary suppression of insulin. Here, we confirm profound diet-PI3K anticancer synergy but show that it is, surprisingly, unrelated to diet macronutrient composition. Instead, the diet-PI3K interaction involves microbiome metabolism of ingested phytochemicals. Specifically, murine ketogenic diet lacks the complex spectrum of phytochemicals found in standard chow, including the soy phytochemicals soyasaponins. We find that soyasaponins are converted by the microbiome into inducers of hepatic cytochrome P450 enzymes, and thereby lower PI3K inhibitor blood levels and anticancer activity. A high-carbohydrate, low-phytochemical diet synergizes with PI3K inhibition to treat cancer in mice, as do antibiotics that curtail the gut microbiome. Thus, diet impacts anticancer drug activity through phytochemical-microbiome-liver interactions.
Collapse
Affiliation(s)
- Asael Roichman
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA
| | - Qianying Zuo
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA; Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Sunghoon Hwang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Wenyun Lu
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA
| | - Ricardo A Cordova
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA
| | - Michael R MacArthur
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA
| | - Jacob A Boyer
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA
| | - Sarah J Mitchell
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA
| | - Jesse Powers
- Rutgers Cancer Institute, New Brunswick, NJ, USA
| | - Sophia A Koval
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Craig J Hunter
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Jamie Rijmers
- Division of Pharmacology, the Netherlands Cancer Institute, Amsterdam 1066 CX, the Netherlands
| | - Rolf-Peter Ryseck
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA
| | - Jenna E AbuSalim
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA; Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Seema Chatterjee
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Won Dong Lee
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Xincheng Xu
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA
| | - Xi Xing
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Zihong Chen
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA
| | - Xianfeng Zeng
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Siddharth Marwaha
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Matthew J McBride
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Jessie Y Guo
- Rutgers Cancer Institute, New Brunswick, NJ, USA
| | - Yibin Kang
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA; Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Mohamed S Donia
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Joshua D Rabinowitz
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA.
| |
Collapse
|
9
|
Peduzzi G, Archibugi L, Farinella R, de Leon Pisani RP, Vodickova L, Vodicka P, Kraja B, Sainz J, Bars-Cortina D, Daniel N, Silvestri R, Uysal-Onganer P, Landi S, Dulińska-Litewka J, Comandatore A, Campa D, Hughes DJ, Rizzato C. The exposome and pancreatic cancer, lifestyle and environmental risk factors for PDAC. Semin Cancer Biol 2025; 113:100-129. [PMID: 40368260 DOI: 10.1016/j.semcancer.2025.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/08/2025] [Accepted: 05/04/2025] [Indexed: 05/16/2025]
Abstract
Pancreatic cancer (PC), particularly pancreatic ductal adenocarcinoma (PDAC), is a significant global health issue with high mortality rates. PDAC, though only 3 % of cancer diagnoses, causes 7 % of cancer deaths due to its severity and asymptomatic early stages. Risk factors include lifestyle choices, environmental exposures, and genetic predispositions. Conditions like new-onset type 2 diabetes and chronic pancreatitis also contribute significantly. Modifiable risk factors include smoking, alcohol consumption, non-alcoholic fatty pancreatic disease (NAFPD), and obesity. Smoking and heavy alcohol consumption increase PC risk, while NAFPD and obesity, particularly central adiposity, contribute through chronic inflammation and insulin resistance. Refined sugar and sugar-sweetened beverages (SSBs) are also linked to increased PC risk, especially among younger individuals. Hormonal treatments and medications like statins, aspirin, and metformin have mixed results on PC risk, with some showing protective effects. The gut microbiome influences PC through the gut-pancreas axis, with disruptions leading to inflammation and carcinogenesis. Exposure to toxic substances, including heavy metals and chemicals, is associated with increased PC risk. Glycome changes, such as abnormal glycosylation patterns, are significant in PDAC development and offer potential for early diagnosis. Interactions between environmental and genetic factors are crucial in PDAC susceptibility. Genome-wide association studies (GWAS) have identified several single nucleotide polymorphisms (SNPs) linked to PDAC, but gene-environment interactions remain largely unexplored. Future research should focus on polygenic risk scores (PRS) and large-scale studies to better understand these interactions and their impact on PDAC risk.
Collapse
Affiliation(s)
| | - Livia Archibugi
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Ruggero Ponz de Leon Pisani
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Ludmila Vodickova
- Biomedical Center Martin, Bioinformatic Center, Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Slovakia; Faculty of Medicine and Biomedical Center in Pilsen, Charles University, Pilsen, Czech Republic
| | - Pavel Vodicka
- Biomedical Center Martin, Bioinformatic Center, Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Slovakia; Faculty of Medicine and Biomedical Center in Pilsen, Charles University, Pilsen, Czech Republic
| | - Bledar Kraja
- University Clinic of Gastrohepatology, University Hospital Center Mother Teresa, Tirana, Albania
| | - Juan Sainz
- Department of Biochemistry and Molecular Biology, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid 28029, Spain; GENYO. Centre for Genomics and Oncological Research. Genomic Oncology department, Granada, Spain; Instituto de Investigación Biosanitaria Ibs.Granada, Granada, Spain
| | - David Bars-Cortina
- Institut Català d'Oncologia (ICO) IDIBELL, Unit of Biomarkers and Susceptibility (UBS), Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain; Institut Català d'Oncologia (ICO) IDIBELL, ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Neil Daniel
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | | | - Pinar Uysal-Onganer
- Cancer Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, London, UK
| | - Stefano Landi
- Department of Biology, University of Pisa, Pisa, Italy
| | | | - Annalisa Comandatore
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | - David J Hughes
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | | |
Collapse
|
10
|
Toriyama K, Masago K, Shibata N, Haneda M, Kuwahara T, Natsume S, Kobayashi S, Fujita Y, Sasaki E, Yamao K, Kawashima H, Shimizu Y, Hara K, Yatabe Y, Hosoda W. Clinicopathological and molecular characterization of KRAS wild-type pancreatic ductal adenocarcinomas reveals precursor lesions with oncogenic mutations and fusions in RAS pathway genes. J Pathol 2025. [PMID: 40317966 DOI: 10.1002/path.6432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 03/07/2025] [Accepted: 03/26/2025] [Indexed: 05/07/2025]
Abstract
Pancreatic ductal adenocarcinomas (PDACs) with wild-type KRAS constitute a small fraction of PDACs, and these tumors were recently shown to harbor frequent actionable oncogenic mutations and fusions. However, the clinicopathological features of KRAS wild-type PDAC have not been well studied. Additionally, precancerous lesions occurring in patients with KRAS wild-type PDACs have rarely been characterized. Here, we investigated the clinicopathological characteristics and outcomes of 75 patients with KRAS wild-type PDAC. Molecular analyses were performed in 40 patients using targeted DNA and whole-exome sequencing and targeted RNA sequencing. We demonstrated that patients with metastatic PDAC with wild-type KRAS were younger (median 59.5 years) than those with mutated KRAS (median 67 years, p < 0.000055). The wild-type KRAS status was not a significant prognostic factor for metastatic disease. Molecularly, genes in the RAS pathway are frequently mutated or rearranged (46%, 16/35), including mutations in BRAF, NRAS, HRAS, EGFR, MAP2K1, FGFR1, FGFR3 and ERBB4 and fusions of FGFR2 (FGFR2::CCDC147, FGFR2::CAT, FGFR2::TXLNA), ALK (STRN::ALK, EML4::ALK), and BRAF (TRIP11::BRAF). Mismatch repair deficiency was identified in 10% (4/39) of patients. Potentially actionable alterations were identified frequently in KRAS wild-type PDACs (30%, 12/40), in which nontubular-type carcinomas were significantly enriched with actionable alterations compared with tubular adenocarcinomas [67% (6/9) versus 16% (5/31); p = 0.007]. Finally, we investigated the precursors of PDACs in 13 pancreatectomy specimens from patients with KRAS wild-type PDAC. We identified three pancreatic intraepithelial neoplasias (PanINs) and two intraductal papillary mucinous neoplasms (IPMNs) harboring oncogenic fusions of ALK and BRAF and driver mutations in BRAF and AKT1. This study suggests that in the context of unmutated KRAS, PDAC is driven by alternative oncogenic mutations or fusions of RAS pathway genes, which may be introduced during the early phase of tumorigenesis. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Kazuhiro Toriyama
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Katsuhiro Masago
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| | - Noriko Shibata
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| | - Masataka Haneda
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| | | | - Seiji Natsume
- Department of Gastroenterological Surgery, Aichi Cancer Center, Nagoya, Japan
| | - Shota Kobayashi
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| | - Yasuko Fujita
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| | - Eiichi Sasaki
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| | - Kenji Yamao
- Department of Gastroenterology, Narita Memorial Hospital, Toyohashi, Japan
| | - Hiroki Kawashima
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Shimizu
- Department of Gastroenterological Surgery, Aichi Cancer Center, Nagoya, Japan
| | - Kazuo Hara
- Department of Gastroenterology, Aichi Cancer Center, Nagoya, Japan
| | - Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Waki Hosoda
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| |
Collapse
|
11
|
Park MK, Lee HJ, Sung JY, Byun HJ, Kim HJ, Kim EJ, Nguyen TM, Kang GJ, Oh SH, Shim JG, Lee H, Nam KT, Kim YY, Rho SB, Kim SG, Lee CH. ERK2-mediated phosphorylation of ZEB1 at S322 enhances PD-L1 expression and EMT, leading to pancreatic cancer progression. Cell Commun Signal 2025; 23:204. [PMID: 40296122 PMCID: PMC12036305 DOI: 10.1186/s12964-025-02182-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 03/29/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Pancreatic cancer is the fourth leading cause of cancer-related deaths. Epithelial-mesenchymal transition (EMT) drives aggressive behaviour and unfavourable outcomes in this disease. The zinc finger E-box-binding homeobox 1 (ZEB1) transcription factor is pivotal in orchestrating EMT, promoting tumor cell mobility, metastasis, and immune evasion through phosphorylation events. However, the precise mechanisms underlying individual phosphorylation sites and their relationship with ZEB1's functions in vivo remain inadequately understood. METHODS We assessed EMT using various techniques, including reverse transcription-quantitative polymerase chain reaction (RT-qPCR), immunoblotting, microscopy, migration, and invasion assays. ZEB1 knockdown was achieved via short hairpin RNA (shRNA), while plasmid transfection facilitated the overexpression of ZEB1, extracellular signal-regulated kinase 1 (ERK1), and extracellular signal-regulated kinase 2 (ERK2). Co-immunoprecipitation and kinase assays were used to examine the interaction between ZEB1 and ERK1/2. PANC-1 and HPAC cells were transplanted in an orthotopic mouse model for in vivo analysis. RESULTS Sphingosylphosphorylcholine (SPC) induced EMT in PANC-1 and HPAC cells in a dose- and time-dependent manner through the phosphorylation and nuclear translocation of ZEB1. Notably, ERK2 interacted with ZEB1 and catalysed the phosphorylation of serine 322 (S322) within the ZEB1 molecule. Disrupting S322 phosphorylation hindered ZEB1's nuclear translocation, leading to reduced programmed death-ligand 1 (PD-L1) expression and suppressed migration and invasion of pancreatic cancer cells. Furthermore, in an orthotopic mouse model, implantation of S322 phosphorylation-deficient (shZEB1/S322A) pancreatic cancer cells suppressed tumour formation and metastasis. We developed a phosphoS322 detection antibody, which validated ZEB1 phosphorylation in pancreatic cancer cells and tissue samples from patients with pancreatic cancer. CONCLUSION SPC induces ZEB1 phosphorylation, with ERK2, rather than ERK1, targeting the S322 site. Inhibiting S322 phosphorylation mitigates EMT, PD-L1 expression, and progression of pancreatic cancer. The phosphoS322 detection antibody might be a valuable tool for predicting pancreatic cancer prognosis. These findings implicate ERK2 as a potential therapeutic target for pancreatic cancer and highlight phosphoZEB1 as a valuable prognostic marker.
Collapse
Affiliation(s)
- Mi Kyung Park
- Department of Biomedical Science, Hwasung Medi-Science University, Hwaseong-si, 18274, Republic of Korea
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 100-715, Goyang, 10326, Republic of Korea
| | - Hye Ja Lee
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 100-715, Goyang, 10326, Republic of Korea
| | - Jee Young Sung
- National Cancer Center, Goyang, 10408, Republic of Korea
| | - Hyun Jung Byun
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 100-715, Goyang, 10326, Republic of Korea
| | - Hyun Ji Kim
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 100-715, Goyang, 10326, Republic of Korea
| | - Eun Ji Kim
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 100-715, Goyang, 10326, Republic of Korea
| | - Tuan Minh Nguyen
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 100-715, Goyang, 10326, Republic of Korea
| | - Gyeoung Jin Kang
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Seung Hyun Oh
- College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jae Gal Shim
- National Cancer Center, Goyang, 10408, Republic of Korea
| | - Ho Lee
- National Cancer Center, Goyang, 10408, Republic of Korea
| | - Ki Taek Nam
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yong Yun Kim
- National Cancer Center, Goyang, 10408, Republic of Korea
| | - Seung Bae Rho
- National Cancer Center, Goyang, 10408, Republic of Korea
| | - Sang Gun Kim
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 100-715, Goyang, 10326, Republic of Korea
| | - Chang Hoon Lee
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Seoul 100-715, Goyang, 10326, Republic of Korea.
| |
Collapse
|
12
|
Roy J, Mouawad R, Kyani A, Hanafi M, Xu Y, Wen B, Sun D, Neamati N. Copper-KRAS-COX2 Axis: A Therapeutic Vulnerability in Pancreatic Cancer. J Med Chem 2025; 68:8400-8428. [PMID: 40135521 DOI: 10.1021/acs.jmedchem.4c03159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
KRAS mutations are a hallmark of pancreatic ductal adenocarcinoma (PDAC), occurring in over 90% of tumors. Tumors with these mutations are highly dependent on copper, making the targeting of copper homeostasis an attractive strategy for treating PDAC due to the higher copper requirement of cancer cells compared to normal cells. Herein, we present the discovery, lead optimization, and structure-activity relationship study of a series of novel quinolyl pyrazinamides for the treatment PDAC. These compounds induce cell death through copper-mediated apoptosis and necroptosis. Our optimized compounds, 39 and 52, are potent, water-soluble and metabolically stable. Compound 52 exhibits 55% oral bioavailability and demonstrates significant in vivo efficacy without obvious toxicity in syngeneic models of PDAC. Additionally, compound 52 showed significant synergy with celecoxib, a selective COX2 inhibitor, both in vitro and in vivo. Our data suggest that compound 52 is a promising candidate for further development in KRAS-mutated cancers.
Collapse
Affiliation(s)
- Joyeeta Roy
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| | - Rima Mouawad
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| | - Armita Kyani
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| | - Maha Hanafi
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Yibin Xu
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, Rogel Cancer Center, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, Rogel Cancer Center, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
13
|
Liu Z, Lenz HJ, Yu J, Zhang L. Differential Response and Resistance to KRAS-Targeted Therapy. Mol Carcinog 2025. [PMID: 40256920 DOI: 10.1002/mc.23908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/10/2025] [Indexed: 04/22/2025]
Abstract
KRAS is the most frequently mutated oncogene. In epithelial malignancies such as lung, colorectal, and pancreatic tumors, KRAS is mutated in 25 to above 90% cases. KRAS was considered undruggable for over three decades until the recent development of covalent inhibitors targeting the KRAS G12C mutant. The recent approval of the KRAS G12C inhibitors sotorasib and adagrasib has ushered in a new era of KRAS-targeted therapy. Despite this success, a major challenge in KRAS-targeted therapy is intrinsic and acquired resistance to KRAS inhibitors. Clinical studies have shown that many patients with KRAS G12C cancers did not respond to sotorasib and adagrasib. Colorectal cancer, in particular, has a markedly lower response rate to KRAS G12C inhibitors compared to non-small cell lung cancer. Furthermore, the therapeutic response to KRAS G12C inhibition was short-lived, with quick emergence of acquired resistance. In this review, we summarize several major themes that have emerged from recent clinical and preclinical studies on the mechanisms of intrinsic and acquired resistance to KRAS-targeted therapy in colorectal, lung, and pancreatic cancers. We also discuss various combination strategies for targeting these mechanisms to overcome resistance to KRAS inhibitors.
Collapse
Affiliation(s)
- Zhaojin Liu
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, California, USA
- Keck School of Medicine of USC, Norris Comprehensive Cancer Center, Los Angeles, California, USA
| | - Heinz-Josef Lenz
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, California, USA
- Keck School of Medicine of USC, Norris Comprehensive Cancer Center, Los Angeles, California, USA
| | - Jian Yu
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, California, USA
- Keck School of Medicine of USC, Norris Comprehensive Cancer Center, Los Angeles, California, USA
| | - Lin Zhang
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, California, USA
- Keck School of Medicine of USC, Norris Comprehensive Cancer Center, Los Angeles, California, USA
| |
Collapse
|
14
|
Cotton K, Comer C, Caporali S, Butera A, Gurres S, Capradossi F, D'Alessandro A, Amelio I, Niklison-Chirou MV. Lipidome atlas of p53 mutant variants in pancreatic cancer. Biol Direct 2025; 20:51. [PMID: 40217553 PMCID: PMC11992884 DOI: 10.1186/s13062-025-00635-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/15/2025] [Indexed: 04/14/2025] Open
Abstract
Mutations in the tumour suppressor protein p53 are present in 70% of human pancreatic ductal adenocarcinomas (PDAC), subsequently to highly common activation mutation of the oncogene KRAS. These p53 mutations generate stable expression of mutant proteins, such as p53R175H and p53R273H, which do not retain p53 wild type function. In this study, we investigated the impact of two specific p53 mutant variants on lipid metabolism of pancreatic cancer. Lipids critically participate to tumorigenesis with to their roles in membrane biosynthesis, energy storage and production of signalling molecules. Using cell lines derived from mouse models of PDAC generated by knock-in p53 alleles carrying point mutations at codons R172H and R270H (equivalent to R175H and R273H in humans), we found that silencing p53R172H and p53R270H in pancreatic cancer cells significantly alters lipid metabolism, with patterns of common and variant specific changes. Specifically, loss of p53R172H in these cells reduces lipid storage. Additionally, silencing either p53R172H or p53R270H individually leads to marked increases in lysophospholipid levels. These findings offer new insights into the lipidome reprogramming induced by the loss of mutant p53 and underscore changes in lipid storage as a potential key molecular mechanism in PDAC pathogenesis.
Collapse
Affiliation(s)
- Kian Cotton
- Life Science Department, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Charley Comer
- Life Science Department, University of Bath, Claverton Down, Bath, BA2 7AY, UK
- Chair for Systems Toxicology, University of Konstanz, Constance, Germany
| | - Sabrina Caporali
- Chair for Systems Toxicology, University of Konstanz, Constance, Germany
| | - Alessio Butera
- Chair for Systems Toxicology, University of Konstanz, Constance, Germany
| | - Stephanie Gurres
- Chair for Systems Toxicology, University of Konstanz, Constance, Germany
| | | | | | - Ivano Amelio
- Chair for Systems Toxicology, University of Konstanz, Constance, Germany.
| | - Maria Victoria Niklison-Chirou
- Life Science Department, University of Bath, Claverton Down, Bath, BA2 7AY, UK.
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
15
|
Hasan R, Zhao Z, Li Y, Liu Y, Zhang Y, Cheng K. Small extracellular vesicles (sEVs) in pancreatic cancer progression and diagnosis. J Control Release 2025; 380:269-282. [PMID: 39889882 PMCID: PMC11908897 DOI: 10.1016/j.jconrel.2025.01.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 02/03/2025]
Abstract
Pancreatic cancer is one of the most aggressive malignancies with poor prognostic outcomes, necessitating the exploration of novel biomarkers and therapeutic targets for early detection and effective treatment. Small extracellular vesicles (sEVs) secreted by cells, have gained considerable attention in cancer research due to their role in intercellular communication and their potential as non-invasive biomarkers. This review focuses on the role of sEVs in the progression of pancreatic cancer and their application as biomarkers. We delve into the biogenesis, composition, and functional implications of sEVs in pancreatic tumor biology, emphasizing their involvement in processes such as tumor growth, metastasis, immune modulation, and chemotherapy resistance. In addition, we discuss the challenges in isolating and characterizing sEVs. The review also highlights recent advances in the utilization of sEV-derived biomarkers for the early diagnosis, prognosis, and monitoring of pancreatic cancer. By synthesizing the latest findings, we aim to underscore the significance of sEVs in pancreatic cancer and their potential to revolutionize patient management through improved diagnostics and targeted therapies.
Collapse
Affiliation(s)
- Reaid Hasan
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Zhen Zhao
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Yuanke Li
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Yanli Liu
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Yuanyuan Zhang
- Institute for Regenerative Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Kun Cheng
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA.
| |
Collapse
|
16
|
Vinaixa J, Martínez-Bosch N, Gibert J, Manero-Rupérez N, Santofimia-Castaño P, Baudou FG, Vera RE, Pease DR, Iglesias M, Sen S, Wang X, Almada LL, Marks DL, Moreno M, Iovanna JL, Rabinovich GA, Fernandez-Zapico ME, Navarro P. Nuclear Galectin-1 promotes KRAS-dependent activation of pancreatic cancer stellate cells. Proc Natl Acad Sci U S A 2025; 122:e2424051122. [PMID: 40172967 PMCID: PMC12002210 DOI: 10.1073/pnas.2424051122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 02/28/2025] [Indexed: 04/04/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive cancers, primarily due to its complex tumor microenvironment (TME), which drives both disease progression and therapy resistance. Understanding the molecular mechanisms governing TME dynamics is essential for developing new treatment strategies for this devastating disease. In this study, we uncover an oncogenic role for Galectin-1 (Gal1), a glycan-binding protein abundantly expressed by activated pancreatic stellate cells (PSCs), a key component of the PDAC TME that orchestrates tumor progression. Our findings reveal that Gal1 expression is elevated in the nucleus of human PSCs in both tissue samples and cultured cell lines. Using chromatin immunoprecipitation followed by sequencing analysis (ChIP-seq), we identify Gal1 occupancy at the promoters of several cancer-associated genes, including KRAS, a pivotal oncogene involved in PDAC pathogenesis. We demonstrate that Gal1 binds to the KRAS promoter, sustaining KRAS expression in PSCs, which, in turn, maintains PSC activation and promotes the secretion of protumorigenic cytokines. Mechanistically, Gal1 is required to preserve histone H3 lysine 4 monomethylation levels and to recruit the histone methyltransferase MLL1 to target promoters. Collectively, our findings define a nuclear function of Gal1 in modulating the transcriptional landscape of cancer-associated genes in PSCs within the PDAC TME, mediated through an epigenetic mechanism. These insights enhance our understanding of PDAC pathology and open potential avenues for therapeutic interventions targeting intracellular Gal1.
Collapse
Affiliation(s)
- Judith Vinaixa
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona08003, Spain
| | - Neus Martínez-Bosch
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona08003, Spain
- Cancer Research Program, Hospital del Mar Research Institute, Associated Unit Hospital del Mar Research Institute/Institute of Biomedical Research of Barcelona-Spanish National Research Council (IIBB-CSIC), Barcelona08003, Spain
| | - Joan Gibert
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona08003, Spain
| | - Noemí Manero-Rupérez
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona08003, Spain
| | - Patricia Santofimia-Castaño
- Translational Research and Innovative Therapies Department, Cancer Research Center of Marseille, INSERM U1068, Institut Paoli-Calmettes, Aix-Marseille University, CNRS, UMR 7258, Marseille13273, France
| | - Federico G. Baudou
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad de Buenos Aires1428, Argentina
- Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján6700, Provincia de Buenos Aires, Argentina
| | - Renzo E. Vera
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN55905
| | - David R. Pease
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN55905
| | - Mar Iglesias
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona08003, Spain
- Departament of Pathology, Hospital del Mar, Barcelona08003, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid28029, Spain
| | - Sandhya Sen
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN55905
| | - Xiyin Wang
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN55905
| | - Luciana L. Almada
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN55905
| | - David L. Marks
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN55905
| | - Mireia Moreno
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona08003, Spain
| | - Juan L. Iovanna
- Translational Research and Innovative Therapies Department, Cancer Research Center of Marseille, INSERM U1068, Institut Paoli-Calmettes, Aix-Marseille University, CNRS, UMR 7258, Marseille13273, France
| | - Gabriel A. Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad de Buenos Aires1428, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires1428, Argentina
- Caixa Research Institute, Barcelona08022, Spain
| | - Martin E. Fernandez-Zapico
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN55905
| | - Pilar Navarro
- Cancer Research Program, Hospital del Mar Research Institute, Associated Unit Hospital del Mar Research Institute/Institute of Biomedical Research of Barcelona-Spanish National Research Council (IIBB-CSIC), Barcelona08003, Spain
- Department of Molecular and Cellular Biomedicine, Institute of Biomedical Research of Barcelona-Spanish National Research Council (IIBB-CSIC), Barcelona08036, Spain
- Institut d’Investigacions Biomediques August Pi Sunyer, Barcelona08036, Spain
| |
Collapse
|
17
|
Stoop TF, Javed AA, Oba A, Koerkamp BG, Seufferlein T, Wilmink JW, Besselink MG. Pancreatic cancer. Lancet 2025; 405:1182-1202. [PMID: 40187844 DOI: 10.1016/s0140-6736(25)00261-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 04/07/2025]
Abstract
Pancreatic cancer is frequently a lethal disease with an aggressive tumour biology often presenting with non-specific symptoms. Median survival is approximately 4 months with a 5-year survival of 13%. Surveillance is recommended in individuals with familial pancreatic cancer, specific mutations, and high-risk intraductal papillary mucinous neoplasm, as they are at high risk of developing pancreatic cancer. Chemotherapy combined with surgical resection remains the cornerstone of treatment. However, only a small subset of patients are candidates for surgery. Multi-agent chemotherapy has improved survival in the palliative setting for patients with metastatic disease, as (neo)adjuvant and induction therapy have in patients with borderline resectable and locally advanced pancreatic. Given that pancreatic cancer is predicted to become the second leading cause of cancer-related death by 2030, novel therapies are urgently needed.
Collapse
Affiliation(s)
- Thomas F Stoop
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Ammar A Javed
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands; Division of Surgical Oncology, Department of Surgery, New York University Medical Center, New York, NY, USA
| | - Atsushi Oba
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan; Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Thomas Seufferlein
- Department of International Medicine I, Ulm University Hospital, Ulm, Germany
| | - Johanna W Wilmink
- Department of Medical Oncology, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Marc G Besselink
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
18
|
Geng C, Zeng J, Deng X, Xia F, Xu X. Molecular Dynamics Investigation into the Stability of KRas and CRaf Multimeric Complexes. J Phys Chem B 2025; 129:3306-3316. [PMID: 40126127 DOI: 10.1021/acs.jpcb.4c08767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
In the Ras/Raf/MAPK signaling pathway, Ras and Raf proteins interact synergistically to form a tetrameric complex. NMR experiments have demonstrated that Ras dimerizes in solution and binds stably to Raf, forming Ras·Raf complexes. In this study, we constructed the ternary and quaternary complexes of KRas and CRaf based on crystal structures, denoted as (KRas)2·CRaf and (KRas)2·(CRaf)2, respectively. Molecular dynamics (MD) simulations were performed to investigate the stability of these complexes, while hydrogen bonds as well as salt bridges formed at the protein-protein interaction interfaces were analyzed based on simulation trajectories. The results revealed that the KRas·CRaf complex is more stable in explicit solvent compared with the KRas dimer. Formation of the stable quaternary complex (KRas)2·(CRaf)2 might be attributed to the association of two binary KRas·CRaf complexes. Additionally, MD simulations of the KRasG12D·CRaf complex revealed a stable and extended binding site at the KRas-CRaf interaction interface. This binding site was identified as a potential therapeutic target to block abnormal signal transmission in the pathway.
Collapse
Affiliation(s)
- Chongli Geng
- School of Chemistry and Molecular Engineering, NYU-ECNU Center for Computational Chemistry at NYU Shanghai, East China Normal University, Shanghai 200062, China
| | - Juan Zeng
- School of Biomedical Engineering, Guangdong Medical University, Dongguan 523808, China
| | - Xianming Deng
- State-province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
- Department of Hematology, The First Affiliated Hospital of Xiamen University, Xiamen University, 361003 Xiamen, China
| | - Fei Xia
- School of Chemistry and Molecular Engineering, NYU-ECNU Center for Computational Chemistry at NYU Shanghai, East China Normal University, Shanghai 200062, China
| | - Xin Xu
- Collaborative Innovation Center of Chemistry for Energy Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, MOE Key Laboratory of Computational Physical Sciences, Department of Chemistry, Fudan University, Shanghai 200433, China
| |
Collapse
|
19
|
Adamopoulos C, Papavassiliou KA, Papavassiliou AG. Targeting the Yin and Yang of KRAS G12C. Trends Mol Med 2025:S1471-4914(25)00058-9. [PMID: 40185675 DOI: 10.1016/j.molmed.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025]
Abstract
In a recent study in Cancer Discovery, Maciag et al. introduce BBO-8520, a novel inhibitor targeting both the active and inactive states of KRASG12C. This dual inhibition shows superior target engagement and prolonged tumor suppression, offering a compelling strategy to overcome resistance development and improve outcomes in KRASG12C-mutant cancers.
Collapse
Affiliation(s)
- Christos Adamopoulos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kostas A Papavassiliou
- First University Department of Respiratory Medicine, Sotiria Chest Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
20
|
Zhou K, Liu Y, Tang C, Zhu H. Pancreatic Cancer: Pathogenesis and Clinical Studies. MedComm (Beijing) 2025; 6:e70162. [PMID: 40182139 PMCID: PMC11965705 DOI: 10.1002/mco2.70162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 03/08/2025] [Accepted: 03/12/2025] [Indexed: 04/05/2025] Open
Abstract
Pancreatic cancer (PC) is a highly lethal malignancy, with pancreatic ductal adenocarcinoma (PDAC) being the most common and aggressive subtype, characterized by late diagnosis, aggressive progression, and resistance to conventional therapies. Despite advances in understanding its pathogenesis, including the identification of common genetic mutations (e.g., KRAS, TP53, CDKN2A, SMAD4) and dysregulated signaling pathways (e.g., KRAS-MAPK, PI3K-AKT, and TGF-β pathways), effective therapeutic strategies remain limited. Current treatment modalities including chemotherapy, targeted therapy, immunotherapy, radiotherapy, and emerging therapies such as antibody-drug conjugates (ADCs), chimeric antigen receptor T (CAR-T) cells, oncolytic viruses (OVs), cancer vaccines, and bispecific antibodies (BsAbs), face significant challenges. This review comprehensively summarizes these treatment approaches, emphasizing their mechanisms, limitations, and potential solutions, to overcome these bottlenecks. By integrating recent advancements and outlining critical challenges, this review aims to provide insights into future directions and guide the development of more effective treatment strategies for PC, with a specific focus on PDAC. Our work underscores the urgency of addressing the unmet needs in PDAC therapy and highlights promising areas for innovation in this field.
Collapse
Affiliation(s)
- Kexun Zhou
- Department of Medical OncologyCancer CenterWest China HospitalSichuan UniversityChengduChina
| | - Yingping Liu
- Department of RadiotherapyCancer HospitalChinese Academy of Medical SciencesBeijingChina
| | - Chuanyun Tang
- The First Clinical Medical College of Nanchang UniversityNanchang UniversityNanchangChina
| | - Hong Zhu
- Department of Medical OncologyCancer CenterWest China HospitalSichuan UniversityChengduChina
- Division of Abdominal Tumor Multimodality TreatmentCancer CenterWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
21
|
Yun D. Letter to the editor regarding "A striking elevation of CA19-9 after preoperative therapy negates prognostic benefit from radical surgery in resectable and borderline resectable pancreatic cancer". Surgery 2025; 180:108850. [PMID: 39396889 DOI: 10.1016/j.surg.2024.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 09/11/2024] [Indexed: 10/15/2024]
Affiliation(s)
- Duo Yun
- Department of Oncology, The First Hospital of Hohhot, Hohhot, China.
| |
Collapse
|
22
|
Cai W, Li Z, Wang W, Liu S, Li Y, Sun X, Sutton R, Deng L, Liu T, Xia Q, Huang W. Resveratrol in animal models of pancreatitis and pancreatic cancer: A systematic review with machine learning. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156538. [PMID: 40037107 DOI: 10.1016/j.phymed.2025.156538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/27/2024] [Accepted: 02/16/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND Resveratrol (RES), a common type of plant polyphenols, has demonstrated promising therapeutic efficacy and safety in animal models of pancreatitis and pancreatic cancer. However, a comprehensive analysis of these data is currently unavailable. This study aimed to systematically review the preclinical evidence regarding RES's effects on animal models of pancreatitis and pancreatic cancer via meta-analyses and optimised machine learning techniques. METHODS Animal studies published from inception until June 30th 2024, were systematically retrieved and manually filtrated across databases including PubMed, EMBASE, Web of Science, Ovid MEDLINE, Scopus, and Cochrane Library. Methodological quality of the included studies was evaluated following the SYRCLE's RoB tool. Predefined outcomes included histopathology and relevant biochemical parameters for acute pancreatitis, and tumour weight/tumour volume for pancreatic cancer, comparing treatment and model groups. Pooled effect sizes of the outcomes were calculated using STATA 17.0 software. Machine learning techniques were employed to predict the optimal usage and dosage of RES in pancreatitis models. RESULTS A total of 50 studies comprising 33 for acute pancreatitis, 1 chronic pancreatitis, and 16 for pancreatic cancer were included for data synthesis after screening 996 records. RES demonstrated significant improvements on pancreatic histopathology score, pancreatic function parameters (serum amylase and lipase), inflammatory markers (TNF-α, IL-1β, IL-6, and pancreatic myeloperoxidase), oxidative biomarkers (malondialdehyde and superoxide dismutase), and lung injury (lung histopathology and myeloperoxidase) in acute pancreatitis models. In pancreatic cancer models, RES notably reduced tumour weight and volume. Machine learning highlighted tree-structured Parzen estimator-optimised gradient boosted decision tree model as achieving the best performance, identifying course after disease induction, total dosage, single dosage, and total number of doses as critical factors for improving pancreatic histology. Optimal single dosage was 20-105 mg/kg with 3 to 9 doses. CONCLUSION This study comprehensively demonstrates the therapeutic effects of RES in mitigating pancreatitis and pancreatic cancer in animal models. Anti-inflammatory, anti-oxidative, and anti-tumour growth properties are potential mechanisms of action for RES.
Collapse
Affiliation(s)
- Wenhao Cai
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ziyu Li
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wen Wang
- Chinese Evidence-based Medicine and Cochrane China Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shiyu Liu
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuying Li
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xin Sun
- Chinese Evidence-based Medicine and Cochrane China Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Robert Sutton
- Liverpool Pancreatitis Research Group, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK
| | - Lihui Deng
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tingting Liu
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Qing Xia
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Wei Huang
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
23
|
Zhang Y, Ma Y, Zhang K, Wang Y, Sun X, Kan C, Han F. KRAS Mutations in Cancer: From Molecular Insights to Therapeutic Strategies. Am J Clin Oncol 2025:00000421-990000000-00275. [PMID: 40167108 DOI: 10.1097/coc.0000000000001192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The global burden of cancer remains a major public health challenge, with Kirsten rat sarcoma viral oncogene homolog (KRAS) emerging as the most common mutated oncogene across diverse malignancies. Once considered "undruggable" due to its unique structure, KRAS has garnered intense research focus, resulting in significant advancements. This paper aims to review recent developments in our understanding of KRAS biology, including its structural and functional aspects, and to explore the latest insights into its mutations across various cancer types. Emphasis is placed on prognosis, predictive roles, and emerging therapeutic strategies targeting KRAS. This review aspires to deepen our comprehension of KRAS and potentially enhance treatment outcomes for cancer patients harboring KRAS mutations in the future.
Collapse
Affiliation(s)
- Yuanzhu Zhang
- Department of Endocrinology and Metabolism, Shandong Provincial Key Medical and Health Discipline of Endocrinology and Laboratory of Endocrinology and Metabolic Diseases, Clinical Research Center
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Yujie Ma
- Department of Endocrinology and Metabolism, Shandong Provincial Key Medical and Health Discipline of Endocrinology and Laboratory of Endocrinology and Metabolic Diseases, Clinical Research Center
| | - Kexin Zhang
- Department of Endocrinology and Metabolism, Shandong Provincial Key Medical and Health Discipline of Endocrinology and Laboratory of Endocrinology and Metabolic Diseases, Clinical Research Center
| | - Yuqun Wang
- Department of Endocrinology and Metabolism, Shandong Provincial Key Medical and Health Discipline of Endocrinology and Laboratory of Endocrinology and Metabolic Diseases, Clinical Research Center
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Shandong Provincial Key Medical and Health Discipline of Endocrinology and Laboratory of Endocrinology and Metabolic Diseases, Clinical Research Center
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Shandong Provincial Key Medical and Health Discipline of Endocrinology and Laboratory of Endocrinology and Metabolic Diseases, Clinical Research Center
| | - Fang Han
- Department of Endocrinology and Metabolism, Shandong Provincial Key Medical and Health Discipline of Endocrinology and Laboratory of Endocrinology and Metabolic Diseases, Clinical Research Center
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| |
Collapse
|
24
|
Su D, Ruan Y, Shi Y, Cao D, Wu T, Dang T, Wang H, Xin Y, Ma M, Meng H, Liu C, Zhang Y. Molecular Subtyping and Genomic Profiling Expand Precision Medicine in KRAS Wild-Type Pancreatic Cancer. Cancer Sci 2025; 116:1094-1106. [PMID: 39833990 PMCID: PMC11967249 DOI: 10.1111/cas.16456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/06/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease with poor prognosis and limited treatment options. While the majority of PDAC cases harbor KRAS mutations, approximately 8%-10% are KRAS wild-type (KRAS-WT). These KRAS-WT tumors often contain actionable mutations and gene fusions, making them more suitable for precision therapies. Identifying these molecular alterations is crucial for improving outcomes in this subset of patients. This retrospective study involved 34 patients with KRAS-WT PDAC. Genomic profiling was performed using next-generation sequencing (NGS) and RNA sequencing to detect mutations and fusions. Comparative analysis was conducted with TCGA-PAAD data, and immune infiltration was assessed using bioinformatic deconvolution methods. Targetable alterations were identified in multiple pathways. Key mutations included ATM (18%), PIK3CA (15%), and ROS1 (15%), while actionable gene fusions such as CCDC6-RET and ETV6-NTRK3 were present in 10.3% of patients. The gene mutations associated with homologous recombination deficiency (HRD) are predicted to increase sensitivity to platinum-based chemotherapy (p = 0.047). Tumors with epigenetic regulatory genes mutations (e.g., ARID1A, KMT2C/D) exhibited enhanced immune cell infiltration, highlighting potential responsiveness to immune checkpoint inhibitors (ICIs). Kinase fusions (NTRK and RET) were linked to response to larotinib and RET-specific inhibitors, respectively. KRAS-WT PDAC contains actionable mutations and fusions, offering significant potential for targeted and immune-based therapies. Further clinical studies are needed to validate these therapeutic approaches.
Collapse
Affiliation(s)
- Dan Su
- Department of Gastrointestinal Medical OncologyHarbin Medical University Cancer HospitalHarbinChina
- Key Laboratory of Tumor Immunology in HeilongjiangHarbinChina
| | - Yuli Ruan
- Department of Gastrointestinal Medical OncologyHarbin Medical University Cancer HospitalHarbinChina
- Key Laboratory of Tumor Immunology in HeilongjiangHarbinChina
- Clinical Research Center for Colorectal Cancer in HeilongjiangHarbinChina
| | - Yingfei Shi
- Department of Translational MedicineGenetron Health (Beijing) Technology, Co. Ltd.BeijingChina
| | - Dandan Cao
- Department of Translational MedicineGenetron Health (Beijing) Technology, Co. Ltd.BeijingChina
| | - Tong Wu
- Department of Gastrointestinal Medical OncologyHarbin Medical University Cancer HospitalHarbinChina
- Key Laboratory of Tumor Immunology in HeilongjiangHarbinChina
- Clinical Research Center for Colorectal Cancer in HeilongjiangHarbinChina
| | - Tianjiao Dang
- Department of Gastrointestinal Medical OncologyHarbin Medical University Cancer HospitalHarbinChina
- Key Laboratory of Tumor Immunology in HeilongjiangHarbinChina
| | - Hong Wang
- Department of Gastrointestinal Medical OncologyHarbin Medical University Cancer HospitalHarbinChina
- Key Laboratory of Tumor Immunology in HeilongjiangHarbinChina
| | - Yaqun Xin
- Department of Translational MedicineGenetron Health (Beijing) Technology, Co. Ltd.BeijingChina
| | - Ming Ma
- Department of Gastrointestinal Medical OncologyHarbin Medical University Cancer HospitalHarbinChina
- Key Laboratory of Tumor Immunology in HeilongjiangHarbinChina
| | - Hongxue Meng
- Department of PathologyHarbin Medical University Cancer HospitalHarbinChina
| | - Chao Liu
- Department of Gastrointestinal Medical OncologyHarbin Medical University Cancer HospitalHarbinChina
- Key Laboratory of Tumor Immunology in HeilongjiangHarbinChina
- Clinical Research Center for Colorectal Cancer in HeilongjiangHarbinChina
| | - Yanqiao Zhang
- Department of Gastrointestinal Medical OncologyHarbin Medical University Cancer HospitalHarbinChina
- Key Laboratory of Tumor Immunology in HeilongjiangHarbinChina
- Clinical Research Center for Colorectal Cancer in HeilongjiangHarbinChina
| |
Collapse
|
25
|
Morand S, Rager L, Craig D, Nemunaitis A, Choucair K, Rao D, Stanbery L, Phinney RC, Walter A, Ghisoli M, Nemunaitis J. Clinical characterization and therapeutic targeting of fusion genes in oncology. Future Oncol 2025; 21:1249-1260. [PMID: 40128124 PMCID: PMC11988278 DOI: 10.1080/14796694.2025.2477974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 03/07/2025] [Indexed: 03/26/2025] Open
Abstract
Gene fusions represent important oncogenic driver mutations resulting in aberrant cellular signaling. In up to 17% of all solid tumors at least one gene fusion can be identified. Precision therapy targeting fusion gene signaling has demonstrated effective clinical benefit. Advancements in clinically relevant next-generation sequencing and bioinformatic techniques have enabled expansion of therapeutic opportunity to subpopulations of patients with fusion gene expression. Clinically, tyrosine inhibitors have shown efficacy in treating fusion gene expressing cancers. Fusion genes are also clonal mutations, meaning it is a personal cancer target involving all cancer cells of that patient, not just a subpopulation of cancer cells within the cancer mass. Thus, both fusion signal disruption and immune signal targeting are effective therapeutic directions. This review discusses fusion gene targeting, therapeutic resistance, and molecular biomarkers.
Collapse
Affiliation(s)
- Susan Morand
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Lauren Rager
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA
| | - Daniel Craig
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA
| | | | - Khalil Choucair
- Department of Hematology/Oncology, Barbara Karmanos Cancer Institute,Wayne State University, Detroit, MI, USA
| | - Donald Rao
- Medical Affairs, Gradalis Inc, Dallas, TX, USA
| | - Laura Stanbery
- Taylor Cancer Research Center, Maumee, OH, USA
- Medical Affairs, Gradalis Inc, Dallas, TX, USA
| | - Richard C. Phinney
- Taylor Cancer Research Center, Maumee, OH, USA
- Department of Hematology/Oncology, Toledo Clinic Cancer Center, Maumee, OH, USA
| | - Adam Walter
- Medical Affairs, Gradalis Inc, Dallas, TX, USA
- Department of Gynecologic Oncology, Promedica Health System, Toledo, OH, USA
| | - Maurizio Ghisoli
- Department of Pediatric Hematology/Oncology, Texas Oncology, P.A, Dallas, TX, USA
| | - John Nemunaitis
- Taylor Cancer Research Center, Maumee, OH, USA
- Medical Affairs, Gradalis Inc, Dallas, TX, USA
- Department of Hematology/Oncology, Toledo Clinic Cancer Center, Maumee, OH, USA
| |
Collapse
|
26
|
Ramesh RPG, Yasmin H, Ponnachan P, Al-Ramadi B, Kishore U, Joseph AM. Phenotypic heterogeneity and tumor immune microenvironment directed therapeutic strategies in pancreatic ductal adenocarcinoma. Front Immunol 2025; 16:1573522. [PMID: 40230862 PMCID: PMC11994623 DOI: 10.3389/fimmu.2025.1573522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 03/04/2025] [Indexed: 04/16/2025] Open
Abstract
Pancreatic cancer is an aggressive tumor with high metastatic potential which leads to decreased survival rate and resistance to chemotherapy and immunotherapy. Nearly 90% of pancreatic cancer comprises pancreatic ductal adenocarcinoma (PDAC). About 80% of diagnoses takes place at the advanced metastatic stage when it is unresectable, which renders chemotherapy regimens ineffective. There is also a dearth of specific biomarkers for early-stage detection. Advances in next generation sequencing and single cell profiling have identified molecular alterations and signatures that play a role in PDAC progression and subtype plasticity. Most chemotherapy regimens have shown only modest survival benefits, and therefore, translational approaches for immunotherapies and combination therapies are urgently required. In this review, we have examined the immunosuppressive and dense stromal network of tumor immune microenvironment with various metabolic and transcriptional changes that underlie the pro-tumorigenic properties in PDAC in terms of phenotypic heterogeneity, plasticity and subtype co-existence. Moreover, the stromal heterogeneity as well as genetic and epigenetic changes that impact PDAC development is discussed. We also review the PDAC interaction with sequestered cellular and humoral components present in the tumor immune microenvironment that modify the outcome of chemotherapy and radiation therapy. Finally, we discuss different therapeutic interventions targeting the tumor immune microenvironment aimed at better prognosis and improved survival in PDAC.
Collapse
Affiliation(s)
- Remya P. G. Ramesh
- Department of Veterinary Medicine, UAE University, Al Ain, United Arab Emirates
| | - Hadida Yasmin
- Immunology and Cell Biology Laboratory, Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal, India
| | - Pretty Ponnachan
- Department of Veterinary Medicine, UAE University, Al Ain, United Arab Emirates
| | - Basel Al-Ramadi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Uday Kishore
- Department of Veterinary Medicine, UAE University, Al Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ann Mary Joseph
- Department of Veterinary Medicine, UAE University, Al Ain, United Arab Emirates
| |
Collapse
|
27
|
Shen J, Wang X, Yu K, Liu K, Wang X, Sun H, Zhou J, Zeng M. Correlation of MRI characteristics with KRAS mutation status in pancreatic ductal adenocarcinoma. Abdom Radiol (NY) 2025:10.1007/s00261-025-04888-x. [PMID: 40156607 DOI: 10.1007/s00261-025-04888-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/26/2025] [Accepted: 03/10/2025] [Indexed: 04/01/2025]
Abstract
PURPOSE To investigate MRI features associated with KRAS mutation status in PDAC and their clinical implications. MATERIALS AND METHODS In our study, 1474 patients pathologically confirmed PDAC patients between January 2016 and December 2023 were evaluated. Patients with genetic testing (KRAS mutation status) and MRI examination were enrolled and grouped as KRAS-mutated PDAC and non-KRAS-mutated PDAC. Contrast-enhanced MRI features, clinicopathologic findings, and prognosis were compared between two groups. RESULTS A total of 308 surgically confirmed PDAC patients (median age, 67 years [IQR, 59, 72]; 183 male and 125 female) with genetic testing data were included, of which 258 had KRAS-mutated PDAC and 50 had non-KRAS-mutated PDAC. KRAS-mutated PDAC demonstrated distinct clinicopathological characteristics, including higher rates of diabetes (OR, 2.450, 95% CI, 1.151-5.212, P = 0.020), pathological peripheral nerve infiltration (OR, 2.296, 95% CI, 1.083-4.867, P = 0.030), and pN stage (OR, 2.006, 95% CI, 1.012-3.976, P = 0.046). The 1-, 3-, 5-year OS rate was worse for KRAS-mutated PDAC (89.9%, 45.4%, 23.2% vs. 95.1%, 60.4% 60.4%, P = 0.045). Rim enhancement (OR = 2.039, 95% CI: 1.053, 3.951, P = 0.035) and larger tumor size (OR = 3.286, 95% CI: 1.523, 7.089, P = 0.002) were identified as distinctive MRI features for KRAS-mutated PDAC. CONCLUSION KRAS-mutated PDAC presents unique clinical and pathological features and is associated with poorer prognosis. Rim enhancement and larger tumor size on MRI were identified as features associated with KRAS-mutated PDAC.
Collapse
Affiliation(s)
- Junjian Shen
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xingxing Wang
- , Department of Pathology, Zhongshan Hospital, Fudan University, PR China
| | - Keqin Yu
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kai Liu
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaolin Wang
- , Department of Interventional Radiology, Zhongshan Hospital, Fudan university, PR China
| | - Haitao Sun
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Jianjun Zhou
- Department of Radiology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, PR China
| | - Mengsu Zeng
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
28
|
Li T, Yu X, Wan X, Liu J, Zheng J, Sun Z, Zhao Y, Chen J, Chen H, Yang Y, Jiang B. Exploiting synthetic lethality in PDAC with antibody drug conjugates and ATR inhibition. Eur J Med Chem 2025; 286:117305. [PMID: 39874630 DOI: 10.1016/j.ejmech.2025.117305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/10/2025] [Accepted: 01/18/2025] [Indexed: 01/30/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a highly lethal malignancy with poor prognosis. Antibody-drug conjugates (ADCs) and their combinations with various anti-tumor drugs have made great progress. Camptothecin, and its derivatives (Dxd, SN-38 or exatecan) targeted TOP1 are effective payloads due to their potent anti-tumor activity. ADCs offer a promising avenue, particularly when integrated with synthetic lethality strategies. In this study, the ADC SA-7-49 is engineered by conjugating exatecan to an anti-TROP2 antibody. The synthetic lethality between camptothecin and the ataxia telangiectasia-mutated and rad3-related (ATR) inhibitors in PDAC cells has been identified through a comprehensive screening of DNA damage response pathways. Drug interactions are quantified using Zero interaction potency (ZIP) scores. RNA sequencing is employed to elucidate the mechanisms driving synergistic effects. ATR inhibitors synergize with camptothecin by inducing apoptosis via ATR-Chk1 pathway inhibition. Knockdown of ATR enhances the sensitivity of PDAC cells to camptothecin and SA-7-49. SA-7-49 selectively targets and eradicates PDAC cells and xenografts without side effects, augmenting anti-tumor activity via synthetic lethality. Our findings reveal a novel therapeutic strategy by integrating ADC technology with synthetic lethality in PDAC.
Collapse
Affiliation(s)
- Tao Li
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Xianqiang Yu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China; School of Physical Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Xinyao Wan
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Jing Liu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Jie Zheng
- School of Information Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Ziyu Sun
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yi Zhao
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Jiakang Chen
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Hongli Chen
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China.
| | - Yifeng Yang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China.
| | - Biao Jiang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
29
|
Liu X, Yang J, Huang S, Hong Y, Zhu Y, Wang J, Wang Y, Liang T, Bai X. Pancreatic cancer-derived extracellular vesicles enhance chemoresistance by delivering KRAS G12D protein to cancer-associated fibroblasts. Mol Ther 2025; 33:1134-1153. [PMID: 39810420 PMCID: PMC11897769 DOI: 10.1016/j.ymthe.2025.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/22/2024] [Accepted: 01/09/2025] [Indexed: 01/16/2025] Open
Abstract
KRAS mutations are instrumental in the development and progression of pancreatic ductal adenocarcinoma (PDAC). Nevertheless, the efficacy of direct targeting of KRAS mutations to inhibit tumor development remains doubtful. It is therefore necessary to gain a deeper insight into the mechanism in which KRAS mutations influence the effectiveness of clinical treatments. In this study, KRASG12D protein was detected in cancer-associated fibroblasts (CAFs) from clinical samples of PDAC. In vitro experiments demonstrated that KRASG12D protein in CAFs was not expressed from its own mutant gene but was derived from the ingestion of tumor cell-derived extracellular vesicles (EVs). The presence of KRASG12D protein in CAFs resulted in enhanced proliferation and migration. Furthermore, KRASG12D-containing CAFs were observed to promote tumor chemoresistance to gemcitabine treatment both in vitro and in vivo. Application of a KRAS mutation-specific inhibitor, MRTX1133, has been demonstrated to reverse chemoresistance in PDAC. Moreover, clinical data suggest that patients with KRAS mutations have poorer prognosis following adjuvant chemotherapy. These findings elucidate the mechanism by which oncogenic KRAS mutations promote cancer chemoresistance and remodel tumor environment in a non-autonomous manner, suggesting a novel strategy for targeting KRAS mutations to enhance chemosensitivity in PDAC.
Collapse
Affiliation(s)
- Xinyuan Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Jiaqi Yang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
| | - Sicong Huang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Yifan Hong
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Yupeng Zhu
- Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Jianing Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Yi Wang
- Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China.
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Pancreatic Disease of Zhejiang Province, Hangzhou, China; Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
30
|
Chaudhary S, Siddiqui JA, Pothuraju R, Bhatia R. Ribosome biogenesis, altered metabolism and ribotoxic stress response in pancreatic ductal adenocarcinoma tumor microenvironment. Cancer Lett 2025; 612:217484. [PMID: 39842499 DOI: 10.1016/j.canlet.2025.217484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy with a poor overall survival rate. Cellular stress response pathways promoting cancer cell fitness in harsh tumor microenvironment (TME) play a critical role in cancer growth and survival. The influence of oncogenic Kras, multi-functional heterogeneous cancer-associated fibroblasts (CAFs), and immunosuppressive TME on cancer cells makes the disease more complex and difficult to treat. The desmoplastic reaction by CAFs comprises approximately 90 % of the tumor, with only 10 % of cancer cells making things even more complicated, resulting in therapy resistance. Consistently increasing fibrosis creates a hypoxic environment and elevated interstitial fluid pressure inside the tumor constraining vascular supply. Stress conditions in TME alter translation efficiency and metabolism to fulfill the energy requirements of rapidly growing cancer cells. Extensive research has been conducted on multiple molecular and metabolic regulators in PDAC TME. However, the role of TME in influencing translation programs, a prerequisite for cell cycle progression and functional/growth requirements for cancer cells, remains elusive. This review highlights the recent advancements in understanding altered translational programs in PDAC TME. We emphasize the role of ribosome biogenesis, ribosome-induced stress response, and the concept of specialized ribosomes and their probable role in mutationally rewiring the pancreatic TME.
Collapse
Affiliation(s)
- Sanjib Chaudhary
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, 781039, Guwahati, Assam, India
| | - Jawed Akhtar Siddiqui
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA; Cancer Center Research Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Ramesh Pothuraju
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram, Kerala, India.
| | - Rakesh Bhatia
- Amity School of Biological Sciences, Amity University Punjab, 82A, Mohali, Punjab, 140306, India.
| |
Collapse
|
31
|
Stukas D, Zievyte I, Ivanauskiene S, Karvelyte G, Jasukaitiene A, Bartkeviciene A, Matthews J, Maimets T, Teino I, Jaudzems K, Gulbinas A, Dambrauskas Z. Small-molecule inhibitor BAY synergizes with gemcitabine through AHR inhibition in pancreatic cancer cells. Biochem Pharmacol 2025; 233:116798. [PMID: 39947435 DOI: 10.1016/j.bcp.2025.116798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/08/2025] [Accepted: 02/10/2025] [Indexed: 02/16/2025]
Abstract
Pancreatic cancer (PC) presents a significant challenge in treatment efficacy due to late-stage diagnosis and chemoresistance. The effects of the combination of a selective small-molecule AHR inhibitor and gemcitabine treatmenteffectiveness in PC cells has been a focus of research. This study utilized the PC cell lines BxPC-3 and Su.86.86 to investigate the impact of AHR activity modulation on gene and protein expression related to the gemcitabine response. Assays including viability measurement, combinational index calculation, qRT-PCR, Western blot analysis, immunocytofluorescence, and clonogenic assays, were employed. Additionally, patient tissue samples were analysed for AHR, ELAVL1, and DCK levels. The results show that AHR activity modulation influenced ELAVL1 localization, DCK expression, and gemcitabine response. Inhibition of AHR activity caused synergistic effects with gemcitabine, whereas activation had an antagonistic effect. Regarding colony formation, inhibition of AHR increased gemcitabine effectiveness by 30-41%, whereas activation decreased the response by 11-28%. Patient tissue analysis revealed correlations between AHR, ELAVL1, and DCK mRNA levels and showed increased levels of AHR protein (2.2-fold) and decreased DCK protein levels (36% decrease) in tumor tissue compared to next-to-cancer tissue. These findings demonstrate the potential of AHR modulation to improve gemcitabine treatment outcomes. This study highlights the significance of AHR modulation in influencing the gemcitabine response in PC cells. By inhibiting AHR activity, cells exhibited improved gemcitabine response, offering a promising avenue for enhancing treatment efficacy. These findings suggest that AHR could serve as a target for optimizing gemcitabine treatment and potentially reducing cancer aggressiveness.
Collapse
Affiliation(s)
- Darius Stukas
- Institute for Digestive Research, Lithuanian University of Health Sciences, Eiveniu 4 50103 Kaunas, Lithuania.
| | - Inga Zievyte
- Institute for Digestive Research, Lithuanian University of Health Sciences, Eiveniu 4 50103 Kaunas, Lithuania.
| | - Sandra Ivanauskiene
- Institute for Digestive Research, Lithuanian University of Health Sciences, Eiveniu 4 50103 Kaunas, Lithuania.
| | - Gabriele Karvelyte
- Institute for Digestive Research, Lithuanian University of Health Sciences, Eiveniu 4 50103 Kaunas, Lithuania.
| | - Aldona Jasukaitiene
- Institute for Digestive Research, Lithuanian University of Health Sciences, Eiveniu 4 50103 Kaunas, Lithuania.
| | - Arenida Bartkeviciene
- Institute for Digestive Research, Lithuanian University of Health Sciences, Eiveniu 4 50103 Kaunas, Lithuania.
| | - Jason Matthews
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, 1046 Blindern 0317 Oslo, Norway; Department of Pharmacology and Toxicology, University of Toronto M5S 1A8 Toronto, Canada.
| | - Toivo Maimets
- Institute of Molecular and Cell Biology, University of Tartu, Riia 23 51010 Tartu, Estonia.
| | - Indrek Teino
- Institute of Molecular and Cell Biology, University of Tartu, Riia 23 51010 Tartu, Estonia.
| | - Kristaps Jaudzems
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006 Latvia.
| | - Antanas Gulbinas
- Institute for Digestive Research, Lithuanian University of Health Sciences, Eiveniu 4 50103 Kaunas, Lithuania
| | - Zilvinas Dambrauskas
- Institute for Digestive Research, Lithuanian University of Health Sciences, Eiveniu 4 50103 Kaunas, Lithuania.
| |
Collapse
|
32
|
Zhao H, Huang Q, Liu YA, Wu W. Oncogenic KRAS Promotes Ferroptosis in Pancreatic Cancer Through Regulation of the Fosl1-Tfrc Axis. Pancreas 2025; 54:e235-e245. [PMID: 39626178 DOI: 10.1097/mpa.0000000000002426] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
ABSTRACT Mutant KRAS activation occurs in most of pancreatic ductal adenocarcinoma (PDAC), which induce the sensitivity to ferroptosis of PDAC cells, but the underlying mechanism is still poorly understood. Here, we show how KRAS acts in signaling to activate transcription factor FOSL1, which promotes the expression of the iron uptake receptor TFRC. In PDAC cells, repression of TFRC by KRAS/FOSL1 signaling inhibited intracellular iron levels, thereby restricting the occurrence of ferroptosis. Furthermore, the KRAS/FOSL1/TFRC axis can make the PDAC cells vulnerable to alteration of the iron level in the tumor microenvironment. Our study highlights a pivotal mechanism of PDAC ferroptosis through iron metabolism and supports a new therapeutic strategy for PDAC with superior potential.
Collapse
Affiliation(s)
- Huijia Zhao
- From the Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | | | | | | |
Collapse
|
33
|
Hu Z, Martí J. Unraveling atomic-scale mechanisms of GDP extraction catalyzed by SOS1 in KRAS-G12 and KRAS-D12 oncogenes. Comput Biol Med 2025; 186:109599. [PMID: 39731920 DOI: 10.1016/j.compbiomed.2024.109599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/30/2024]
Abstract
The guanine exchange factor SOS1 plays a pivotal role in the positive feedback regulation of the KRAS signaling pathway. Recently, the regulation of KRAS-SOS1 interactions and KRAS downstream effector proteins has emerged as a key focus in the development of therapies targeting KRAS-driven cancers. However, the detailed dynamic mechanisms underlying SOS1-catalyzed GDP extraction and the impact of KRAS mutations remain largely unexplored. In this study, we unveil and describe in atomic detail the primary mechanisms by which SOS1 facilitates GDP extraction from KRAS oncogenes. For GDP-bound wild-type KRAS (KRAS-G12), four critical amino acids (Lys811, Glu812, Lys939, and Glu942) are identified as essential for the catalytic function of SOS1. Notably, the KRAS-G12D mutation (KRAS-D12) significantly accelerates the rate of GDP extraction. The molecular basis of this enhancement are attributed to hydrogen bonding interactions between the mutant residue Asp12 and a positively charged pocket in the intrinsically disordered region (residues 807-818), comprising Ser807, Trp809, Thr810, and Lys811. These findings provide novel insights into SOS1-KRAS interactions and offer a foundation for developing anti-cancer strategies aimed at disrupting these mechanisms.
Collapse
Affiliation(s)
- Zheyao Hu
- Department of Physics, Polytechnic University of Catalonia-Barcelona Tech, B4-B5 Northern Campus UPC, Barcelona, 08034, Catalonia, Spain
| | - Jordi Martí
- Department of Physics, Polytechnic University of Catalonia-Barcelona Tech, B4-B5 Northern Campus UPC, Barcelona, 08034, Catalonia, Spain.
| |
Collapse
|
34
|
Ma R, Li G, Ye Y, Liang L, Wang C, Zhou H, Zhang P, An L, Shi G, Chen Q, Xu H, Gao Z. Prognosis conferred by molecular features of appendix-derived Pseudomyxoma Peritonei. Transl Oncol 2025; 53:102279. [PMID: 39929063 DOI: 10.1016/j.tranon.2025.102279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/05/2025] [Indexed: 03/01/2025] Open
Abstract
INTRODUCTION Pseudomyxoma Peritonei (PMP) is an extremely rare disease characterized by progressive accumulation of mucinous ascites and implants in the peritoneum. We investigated the prognostic value for response to cytoreductive surgery (CRS) or hyperthermic intraperitoneal chemotherapy (HIPEC) and dissected potential beneficial targeted therapy utilizing genomic characteristics. METHODS Whole-exome sequencing (WES) was performed on tissue specimens and matched white blood cells from 81 patients with PMP. The study investigated mutational signatures, profiling, and their correlation with progression-free survival (PFS) and overall survival (OS). RESULTS Signature 3 (HRD) and signature 15 (dMMR) were dominant. NMF cluster 1, characterized by signature 4, exhibited a worse prognosis. The p53 and TGF-β signaling pathways may contribute as risk factors for worse OS and PFS, respectively. MUC16-mutated patients had worse PFS (P = 0.016) and OS (P = 0.004) compared to wild-type patients. Patients with tumor mutational burden (TMB) > 1(P = 0.026) or alterations in TP53 (P = 0.006) or SMAD4 (P = 0.013) had significantly worse OS compared to those with a TMB < 1 or normal genes. Patients with homologous recombination deficiency (HRD) positivity (P = 0.003) or alterations in TGFBR2 (P = 0.037) experienced worse PFS compared to their respective control groups. Furthermore, NMF cluster1 (P = 0.020), TP53 (P = 0.004), and MUC16 (P = 0.013) were identified as independent prognostic factors for OS, while HRD status (P = 0.003) was independent predictors for PFS in PMP. CONCLUSIONS The study reveals that genomic profiling can serve as a robust tool for identifying prognostic markers in PMP. The identified genomic mutations and signaling pathway offer new avenues for targeted therapies.
Collapse
Affiliation(s)
- Ruiqing Ma
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China; Department of Myxoma, Aerospace Center Hospital, Beijing, 100049, China.
| | - Guojun Li
- Thorgene Co., Ltd., Beijing, 100176, China.
| | - Yingjiang Ye
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China.
| | - Lei Liang
- Department of Ultrasound, Aerospace Center Hospital, Beijing, China
| | - Chong Wang
- Department of Myxoma, Aerospace Center Hospital, Beijing, 100049, China
| | - Haipeng Zhou
- Department of Myxoma, Aerospace Center Hospital, Beijing, 100049, China
| | - Pu Zhang
- Department of Myxoma, Aerospace Center Hospital, Beijing, 100049, China
| | - Lubiao An
- Department of Myxoma, Aerospace Center Hospital, Beijing, 100049, China
| | - Guanjun Shi
- Department of Myxoma, Aerospace Center Hospital, Beijing, 100049, China
| | - Qian Chen
- Thorgene Co., Ltd., Beijing, 100176, China.
| | - Hongbin Xu
- Department of Myxoma, Aerospace Center Hospital, Beijing, 100049, China.
| | - Zhidong Gao
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
35
|
Daya T, Breytenbach A, Gu L, Kaur M. Cholesterol metabolism in pancreatic cancer and associated therapeutic strategies. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159578. [PMID: 39542394 DOI: 10.1016/j.bbalip.2024.159578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/31/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
Pancreatic cancer remains one of the most lethal cancers due to late diagnosis and high chemoresistance. Despite recent progression in the development of chemotherapies, immunotherapies, and potential nanoparticles-based approaches, the success rate of therapeutic response is limited which is further compounded by cancer drug resistance. Understanding of emerging biological and molecular pathways causative of pancreatic cancer's aggressive and chemoresistance is vital to improve the effectiveness of existing therapeutics and to develop new therapies. One such under-investigated and relatively less explored area of research is documenting the effect that lipids, specifically cholesterol, and its metabolism, impose on pancreatic cancer. Dysregulated cholesterol metabolism has a profound role in supporting cellular proliferation, survival, and promoting chemoresistance and this has been well established in various other cancers. Thus, we aimed to provide an in-depth review focusing on the significance of cholesterol metabolism in pancreatic cancer and relevant genes at play, molecular processes contributing to cellular cholesterol homeostasis, and current research efforts to develop new cholesterol-targeting therapeutics. We highlight the caveats, weigh in different experimental therapeutic strategies, and provide possible suggestions for future research highlighting cholesterol's importance as a therapeutic target against pancreatic cancer resistance and cancer progression.
Collapse
Affiliation(s)
- Tasvi Daya
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa
| | - Andrea Breytenbach
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa
| | - Liang Gu
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa
| | - Mandeep Kaur
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa.
| |
Collapse
|
36
|
Nan K, Zhang L, Zou Y, Geng Z, Huang J, Peng Y, Yin S, Zhang M. Integrated Profiling Delineated KIF18A as a Significant Biomarker Associated with Both Prognostic Outcomes and Immune Response in Pancreatic Cancer. Immunotargets Ther 2025; 14:123-138. [PMID: 40040635 PMCID: PMC11878147 DOI: 10.2147/itt.s497284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/08/2025] [Indexed: 03/06/2025] Open
Abstract
Purpose Kinesin family member 18A (KIF18A) is a member of the kinesin-8 family of motor proteins, involved in the progression and metastasis of various tumors. However, its role in pancreatic adenocarcinoma (PAAD) remains unclear. Methods To evaluate that role, RNA sequencing datasets, complemented by pertinent clinical metadata, were procured from the Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) repositories. The protein expression level of KIF18A in PAAD was derived from human protein atlas (HPA) database. The differences in KIF18A expression levels and prognostic related genes were identified through multivariate Cox regression and Lasso regression analysis to construct a prognostic risk model. The Tumor Mutation Burden (TMB), Microsatellite (MSI), immune landscape, mutation landscape and drug sensitivity of high- and low-expression KIF18A groups were assessed in immunotherapy cohorts and KIF18A expression cohorts. Finally, in vitro experiments were conducted to elucidate the molecular function of KIF18A in regulating the malignant behavior of PAAD. Results KIF18A is highly expressed in PAAD and is closely related to worse clinical stage and poor prognosis. Single cell analysis revealed that KIF18A is mainly expressed in microtubules of tumor cells and participated in mitosis and cell cycle of PAAD. Further analysis revealed that the expression of KIF18A is closely related to TMB, MSI, and immune cell infiltration. In vitro experiments confirmed that KIF18A promotes the proliferation, migration and expression of adhesion molecules in PAAD, and inhibits angiogenesis. In addition, the high expression of KIF18A is positively related to ferroptosis and m6A genes expression, and its high expression is driven by mutated KRAS and TP53. Conclusion This study confirmed that KIF18A can be used as a marker to predict the prognosis and immunotherapy of PAAD, and it participates in the formation of microtubules in PAAD cells and promotes the malignant behavior of PAAD.
Collapse
Affiliation(s)
- Kai Nan
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi, 710054, People’s Republic of China
| | - Lei Zhang
- Department of Pharmacy, Shaanxi Provincial Hospital of Chinese Medicine, Xi’an, Shaanxi, 710003, People’s Republic of China
| | - Yujia Zou
- Department of Orthopaedics, The second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710000, People’s Republic of China
| | - Zilong Geng
- Department of Orthopaedics, The second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710000, People’s Republic of China
| | - Jing Huang
- Department of Rehabilitative Medicine, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, People’s Republic of China
| | - Yulong Peng
- Department of Rehabilitative Medicine, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, People’s Republic of China
| | - Su Yin
- Department of Rehabilitative Medicine, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, People’s Republic of China
| | - Ming Zhang
- Department of General Practice, Honghui Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi, 710054, People’s Republic of China
| |
Collapse
|
37
|
Charette M, Rosenblum C, Shade O, Deiters A. Optogenetics with Atomic Precision─A Comprehensive Review of Optical Control of Protein Function through Genetic Code Expansion. Chem Rev 2025; 125:1663-1717. [PMID: 39928721 PMCID: PMC11869211 DOI: 10.1021/acs.chemrev.4c00224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 02/12/2025]
Abstract
Conditional control of protein activity is important in order to elucidate the particular functions and interactions of proteins, their regulators, and their substrates, as well as their impact on the behavior of a cell or organism. Optical control provides a perhaps optimal means of introducing spatiotemporal control over protein function as it allows for tunable, rapid, and noninvasive activation of protein activity in its native environment. One method of introducing optical control over protein activity is through the introduction of photocaged and photoswitchable noncanonical amino acids (ncAAs) through genetic code expansion in cells and animals. Genetic incorporation of photoactive ncAAs at key residues in a protein provides a tool for optical activation, or sometimes deactivation, of protein activity. Importantly, the incorporation site can typically be rationally selected based on structural, mechanistic, or computational information. In this review, we comprehensively summarize the applications of photocaged lysine, tyrosine, cysteine, serine, histidine, glutamate, and aspartate derivatives, as well as photoswitchable phenylalanine analogues. The extensive and diverse list of proteins that have been placed under optical control demonstrates the broad applicability of this methodology.
Collapse
Affiliation(s)
- Maura Charette
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Carolyn Rosenblum
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Olivia Shade
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Alexander Deiters
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
38
|
Hong Z, Huang X, Xia L, Liang T, Bai X. Reciprocal regulation of MMP-28 and EGFR is required for sustaining proliferative signaling in PDAC. J Exp Clin Cancer Res 2025; 44:68. [PMID: 39994761 PMCID: PMC11849219 DOI: 10.1186/s13046-025-03323-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/06/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUD Sustaining proliferation signaling is the top hallmarks of cancer, driving continuous tumor growth and resistance to drug treatments. Blocking proliferation signaling has shown limited benefit in clinical treatment of pancreatic ductal adenocarcinoma, highlighting the urgent need to deeply understand proliferation signaling and develop new therapeutic strategies. METHODS By leveraging clinical data and data from the TCGA and GDSC datasets, we investigated the association between MMP-28 expression and the sensitivity to EGFR inhibitors as well as the prognosis of PDAC. Transcriptomic and biological experiments explore the regulatory role of MMP-28 on the EGFR signaling pathway. Additionally, in vitro and in vivo studies are employed to evaluate MMP-28 as a biomarker for sensitivity to EGFR inhibitors. RESULTS We found that MMP-28, a metalloproteinase, was significantly associated with the sensitivity to EGFR inhibitors. Furthermore, MMP-28 could promote PDAC growth and metastasis. Mechanistically, MMP-28 facilitated the maturation and release of the TGF-α precursor, thus promoting EGFR activation. In return, EGFR upregulated MMP-28 through AP-1-mediated transcription, forming a positive feedback loop that provided sustaining proliferation signaling for PDAC. Subsequently, MMP-28 was identified to predict the response to EGFR inhibitors and recognize responsive patients. CONCLUSIONS Our findings revealed the role of MMP-28 and EGFR in generation of sustaining proliferation signaling and provided a new therapy strategy for PDAC.
Collapse
Affiliation(s)
- Zhengtao Hong
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xing Huang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- Zhejiang University Cancer Center, Hangzhou, 310063, China.
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, 310003, China.
| | - Linghao Xia
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Zhejiang Province, Hangzhou, 31003, China.
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China.
- Zhejiang University Cancer Center, Hangzhou, 310063, China.
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, 310003, China.
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Zhejiang Province, Hangzhou, 31003, China.
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China.
- Zhejiang University Cancer Center, Hangzhou, 310063, China.
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, 310003, China.
| |
Collapse
|
39
|
Sobhani N, Pittacolo M, D’Angelo A, Marchegiani G. Recent Anti-KRAS G12D Therapies: A "Possible Impossibility" for Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2025; 17:704. [PMID: 40002297 PMCID: PMC11853620 DOI: 10.3390/cancers17040704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/13/2025] [Accepted: 02/16/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer, able to thrive in a challenging tumor microenvironment. Current standard therapies, including surgery, radiation, chemotherapy, and chemoradiation, have shown a dismal survival prognosis, resulting in less than a year of life in the metastatic setting. Methods: The pressing need to find better therapeutic methods brought about the discovery of new targeted therapies against the infamous KRAS mutations, the major oncological drivers of PDAC. Results: The most common KRAS mutation is KRASG12D, which causes a conformational change in the protein that constitutively activates downstream signaling pathways driving cancer hallmarks. Novel anti-KRASG12D therapies have been developed for solid-organ tumors, including small compounds, pan-RAS inhibitors, protease inhibitors, chimeric T cell receptors, and therapeutic vaccines. Conclusions: This comprehensive review summarizes current knowledge on the biology of KRAS-driven PDAC, the latest therapeutic options that have been experimentally validated, and developments in ongoing clinical trials.
Collapse
Affiliation(s)
- Navid Sobhani
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Matteo Pittacolo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35122 Padova, Italy;
| | - Alberto D’Angelo
- Department of Medicine, Northern General Hospital, Sheffield S5 7AT, UK;
| | - Giovanni Marchegiani
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35122 Padova, Italy;
| |
Collapse
|
40
|
Jiang X, Wang T, Zhao B, Sun H, Dong Y, Ma Y, Li Z, Wu Y, Wang K, Guan X, Long B, Qin L, Shi W, Shi L, He Q, Liu W, Li M, Xiao L, Zhou C, Sun H, Yang J, Guan J, Zhou H, Yu Z, Jiao Z. KRAS G12D-driven pentose phosphate pathway remodeling imparts a targetable vulnerability synergizing with MRTX1133 for durable remissions in PDAC. Cell Rep Med 2025; 6:101966. [PMID: 39970873 PMCID: PMC11866490 DOI: 10.1016/j.xcrm.2025.101966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 11/04/2024] [Accepted: 01/16/2025] [Indexed: 02/21/2025]
Abstract
The KRASG12D inhibitor MRTX1133 shows the potential to revolutionize the treatment paradigm for pancreatic ductal adenocarcinoma (PDAC), yet presents challenges. Our findings indicate that KRASG12D remodels a pentose phosphate pathway (PPP)-dominant central carbon metabolism pattern, facilitating malignant progression and resistance to MRTX1133 in PDAC. Mechanistically, KRASG12D drives excessive degradation of p53 and glucose-6-phosphate dehydrogenase (G6PD)-mediated PPP reprogramming through retinoblastoma (Rb)/E2F1/p53 axis-regulated feedback loops that amplify ubiquitin-conjugating enzyme E2T (UBE2T) transcription. Genetic ablation or pharmacological inhibition of UBE2T significantly suppresses PDAC progression and potentiates MRTX1133 efficacy. Leveraging structure advantages of the UBE2T inhibitor pentagalloylglucose (PGG), we develop a self-assembling nano co-delivery system with F-127, PGG, and MRTX1133. This system enhances the efficacy of PGG and MRTX1133, achieving durable remissions (85% overall response rate) and long-term survival (100% progression-free survival) in patient-derived xenografts and spontaneous PDAC mice. This study reveals the role of KRASG12D-preferred PPP reprogramming in MRTX1133 resistance and proposes a potentially therapeutic strategy for KRASG12D-mutated PDAC.
Collapse
Affiliation(s)
- Xiangyan Jiang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Tao Wang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Bin Zhao
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Haonan Sun
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Yuman Dong
- Gansu Province High-Altitude High-Incidence Cancer Biobank, Lanzhou University Second Hospital, Lanzhou 730000, China; Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730000, China; State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou 730000, China
| | - Yong Ma
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Zhigang Li
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Yuxia Wu
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Keshen Wang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Xiaoying Guan
- Department of Pathology, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Bo Long
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Long Qin
- Gansu Province High-Altitude High-Incidence Cancer Biobank, Lanzhou University Second Hospital, Lanzhou 730000, China; Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Wengui Shi
- Gansu Province High-Altitude High-Incidence Cancer Biobank, Lanzhou University Second Hospital, Lanzhou 730000, China; Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Lei Shi
- School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Qichen He
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Wenbo Liu
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Mingdou Li
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Lixia Xiao
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730000, China
| | - Chengliang Zhou
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
| | - Hui Sun
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Jing Yang
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Junhong Guan
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Huinian Zhou
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Zeyuan Yu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; Gansu Province High-Altitude High-Incidence Cancer Biobank, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Zuoyi Jiao
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou 730000, China; Gansu Province High-Altitude High-Incidence Cancer Biobank, Lanzhou University Second Hospital, Lanzhou 730000, China.
| |
Collapse
|
41
|
Lena J, Alamé M, Italiano A, Soubeyran I, Blouin L, Khalifa E, Cousin S, Pernot S, Palmieri LJ. Extensive molecular profiling of KRAS wild-type as compared to KRAS mutated pancreatic ductal adenocarcinoma on 318 patients. Eur J Cancer 2025; 216:115197. [PMID: 39729677 DOI: 10.1016/j.ejca.2024.115197] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/15/2024] [Accepted: 12/18/2024] [Indexed: 12/29/2024]
Abstract
PURPOSE Molecular profiling is increasingly implemented to guide treatment of advanced pancreatic ductal adenocarcinoma (PDAC), especially when for clinical trials enrollment. This study aimed to describe actionable alterations detected in KRAS mutated (KRASm) versus KRAS wild-type (KRASwt) PDAC, the latter group being considered enriched in molecular alterations. METHODS This prospective monocentric study included patients with locally advanced or metastatic PDAC who underwent next-generation sequencing (NGS) on liquid biopsy and/or tissue samples between 2015 and 2023, as part of the BIP academic study (NCT02534649). Actionable alterations were classified using the ESCAT (ESMO Scale for Clinical Actionability of molecular Targets). RESULTS A total of 378 patients with a PDAC underwent NGS: 73 on tissue samples, 162 on liquid biopsies, and 143 on both tissue and liquid. Liquid biopsies had a 59.3 % performance (181 informative samples out of 305). Among 318 informative NGS samples, 273 (86 %) were KRASm, and 45 (14 %) were KRASwt. Median overall survival (OS) was 19.35 in KRASwt patients and 16.89 months for KRASm patients (HR 0.67, 95 %CI (0.49-0.90), p = 0.02). ESCAT alterations were found in 15.7 % of total population, with 31.1 % in KRASwt tumors and 13.2 % in KRASm tumors. BRCA1/2 mutations were identified in 7.5 % of the population, and one NTRK fusion was found in a KRASwt PDAC. The molecular tumor board considered 71 patients (22.3 %) eligible for early-phase trials, with 14 treated with matched therapy. CONCLUSION Although actionable mutations were more frequent in KRASwt tumors, 13.2 % of KRASm PDAC harbored ESCAT alterations, emphasizing the importance of molecular profiling regardless of KRAS status.
Collapse
Affiliation(s)
- Jeanne Lena
- Department of Medicine, Institut Bergonié, Bordeaux, France
| | - Mélissa Alamé
- Department of Pathology, Institut Bergonié, Bordeaux, France
| | - Antoine Italiano
- Department of Medicine, Institut Bergonié, Bordeaux, France; Faculty of Medicine, University of Bordeaux, France
| | | | - Laura Blouin
- Department of Pathology, Institut Bergonié, Bordeaux, France
| | | | - Sophie Cousin
- Department of Medicine, Institut Bergonié, Bordeaux, France
| | - Simon Pernot
- Department of Medicine, Institut Bergonié, Bordeaux, France
| | | |
Collapse
|
42
|
Qiao C, Wang L, Huang C, Jia Q, Bao W, Guo P, Tan D, Chen Z, Shi C, Rao Z, Zhang R, Wei W, Wang Z. Engineered Bacteria Manipulate Cysteine Metabolism to Boost Ferroptosis-Based Pancreatic Ductal Adenocarcinoma Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2412982. [PMID: 39696900 DOI: 10.1002/adma.202412982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/18/2024] [Indexed: 12/20/2024]
Abstract
Cysteine metabolism is a key determinant of the defense against ferroptosis in pancreatic ductal adenocarcinoma (PDAC). Blocking cysteine metabolism may trigger potent ferroptosis in PDAC cells by generating lipid peroxides during tumor metabolic processes. However, current methods to limit cysteine availability fall short, failing to efficiently block cysteine metabolism due to inadequate tumor targeting and compensatory cysteine sources. Inspired by sulfur-metabolizing bacteria, synthetic biology to develop an engineered bacterium capable of directly depleting cysteine to block its metabolism is used. Acting as a living drug, these engineered bacteria colonize the tumor and continuously produce engineered cyst(e)inase enzyme (CGL) under the stimulation of tumor hypoxia. The CGL exhausts the substrate cysteine, completely impeding cysteine metabolism. This process dismantles the ferroptosis defense system in PDAC cells, triggers potent ferroptosis, and achieves efficient treatment. The results demonstrate that engineered bacteria designed for cysteine metabolism modulation possess unparalleled advantages in efficacy, persistence, and precision in blocking cysteine metabolism, making them highly suitable for effective ferroptosis treatment of PDAC.
Collapse
Affiliation(s)
- Chaoqiang Qiao
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, 710126, P. R. China
- Guangzhou Institute of Technology, Xidian University, Guangzhou, 510555, P. R. China
| | - Lexuan Wang
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, 710126, P. R. China
| | - Chuting Huang
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, 710126, P. R. China
| | - Qian Jia
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, 710126, P. R. China
| | - Weier Bao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Peilin Guo
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Dengxu Tan
- Laboratory Animal Center, Air Force Medical University, Xi'an, 710032, P. R. China
| | - Zhuang Chen
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, 710126, P. R. China
| | - Changhong Shi
- Laboratory Animal Center, Air Force Medical University, Xi'an, 710032, P. R. China
| | - Zhiping Rao
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, 710126, P. R. China
| | - Ruili Zhang
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, 710126, P. R. China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zhongliang Wang
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, 710126, P. R. China
| |
Collapse
|
43
|
Jin D, Khan NU, Gu W, Lei H, Goel A, Chen T. Informatics strategies for early detection and risk mitigation in pancreatic cancer patients. Neoplasia 2025; 60:101129. [PMID: 39842383 PMCID: PMC11763847 DOI: 10.1016/j.neo.2025.101129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/17/2025] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
This review provides a comprehensive overview of the current landscape in pancreatic cancer (PC) screening, diagnosis, and early detection. This emphasizes the need for targeted screening in high-risk groups, particularly those with familial predispositions and genetic mutations, such as BRCA1, BRCA2, and PALB2. This review highlights the sporadic nature of most PC cases and significant risk factors, including smoking, alcohol consumption, obesity, and diabetes. Advanced imaging techniques, such as Endoscopic Ultrasound (EUS) and Contrast-Enhanced Harmonic Imaging (CEH-EUS), have been discussed for their superior sensitivity in early detection. This review also explores the potential of novel biomarkers, including those found in body fluids, such as serum, plasma, urine, and bile, as well as the emerging role of liquid biopsy technologies in analyzing circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and exosomes. AI-driven approaches, such as those employed in Project Felix and CancerSEEK, have been highlighted for their potential to enhance early detection through deep learning and biomarker discovery. This review underscores the importance of universal genetic testing and the integration of AI with traditional diagnostic methods to improve outcomes in high-risk individuals. Additionally, this review points to future directions in PC diagnostics, including next-generation imaging, molecular biomarkers, and personalized medicine, aiming to overcome current diagnostic challenges and improve survival rates. Ultimately, the review advocates the adoption of informatics and AI-driven strategies to enhance early detection, reduce morbidity, and save lives in the fight against pancreatic cancer.
Collapse
Affiliation(s)
- Di Jin
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310018, China; Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Najeeb Ullah Khan
- Institute of Biotechnology & Genetic Engineering (Health Division), The University of Agriculture Peshawar, Peshawar, PO Box 25130, Pakistan.
| | - Wei Gu
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310018, China; Wenzhou Medical University, Wenzhou, 325000, China.
| | - Huijun Lei
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310018, China.
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, California, USA; City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| | - Tianhui Chen
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310018, China.
| |
Collapse
|
44
|
Li Z, Wang D, Zhu X. Unveiling the functions of five recently characterized lncRNAs in cancer progression. Clin Transl Oncol 2025; 27:458-465. [PMID: 39066874 DOI: 10.1007/s12094-024-03619-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/11/2024] [Indexed: 07/30/2024]
Abstract
Numerous studies over the past few decades have shown that RNAs are multifaceted, multifunctional regulators of most cellular processes, contrary to the initial belief that they only act as mediators for translating DNA into proteins. LncRNAs, which refer to transcripts longer than 200nt and lack the ability to code for proteins, have recently been identified as central regulators of a variety of biochemical and cellular processes, particularly cancer. When they are abnormally expressed, they are closely associated with tumor occurrence, metastasis, and tumor staging. Therefore, through searches on Google Scholar, PubMed, and CNKI, we identified five five recently characterized lncRNAs-Lnc-SLC2A12-10:1, LncRNA BCRT1, lncRNA IGFBP4-1, LncRNA PCNAP1, and LncRNA CDC6-that have been linked to the promotion of cancer cell proliferation, invasion, and metastasis. Consequently, this review encapsulates the existing research and molecular underpinnings of these five newly identified lncRNAs across various types of cancer. It suggests that these novel lncRNAs hold potential as independent biomarkers for clinical diagnosis and prognosis, as well as candidates for therapeutic intervention. In parallel, we discuss the challenges inherent in the research on these five newly discovered lncRNAs and look forward to the avenues for future exploration in this field.
Collapse
Affiliation(s)
- Zhicheng Li
- Department of Urology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| | - Dan Wang
- Department of Urology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China
| | - Xiaojun Zhu
- Department of Urology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, China.
| |
Collapse
|
45
|
Wu H, Xie Y, Li A, Liu X, Guo L, Wu F, Yang Z, Zhang Z, Zhang X. LncRNA RNF144A-AS1 gene polymorphisms and their influence on lung cancer patients in the Chinese Han population. Noncoding RNA Res 2025; 10:252-260. [PMID: 39611047 PMCID: PMC11602538 DOI: 10.1016/j.ncrna.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/16/2024] [Accepted: 10/30/2024] [Indexed: 11/30/2024] Open
Abstract
Lung cancer is primarily classified as NSCLC, which is distinguished by a wide range of genetic variations. This study focused on RNF144A-AS1, a relatively unexplored lncRNA, to explore the impact of its genetic polymorphisms on the susceptibility to NSCLC. We detected RNF144A-AS1 expression and its correlation with prognosis and clinical pathological features using bioinformatics analysis. The association between RNF144A-AS1 polymorphism and NSCLC susceptibility was evaluated using case-control methods. This investigation featured a cohort of 700 NSCLC individuals and 700 healthy controls. The genotype of genetic variation was detected by PCR-RFLP and iMLDR, followed by subsequent calculation of OR and 95 % CI. Our data show that RNF144A-AS1 exhibits high expression levels in LUAD tissues and its expression is closely linked to LUAD progression and prognosis. Carrier of RNF144A-AS1 rs3806609 TT genotype increased NSCLC susceptibility compared to carrier of rs3806609 CC genotype (OR = 2.21, 95%CI = 1.57-3.13). Our study identifies RNF144A-AS1 genetic variants as potential susceptibility markers in NSCLC. RNF144A-AS1 promotes cell proliferation and migration in LUAD through the IFN-γ/JAK2/STAT1 signalling pathway. Collectively, these findings pave the way for developing targeted therapies and diagnostic tools based on RNF144A-AS1 and its variants.
Collapse
Affiliation(s)
- Hongjiao Wu
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China
- College of Life Science, North China University of Science and Technology, Tangshan, 063210, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, Tangshan, 063210, China
| | - Yuning Xie
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China
| | - Ang Li
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China
| | - Xiyao Liu
- College of Life Science, North China University of Science and Technology, Tangshan, 063210, China
| | - Liwen Guo
- College of Life Science, North China University of Science and Technology, Tangshan, 063210, China
| | - Fengjun Wu
- College of Life Science, North China University of Science and Technology, Tangshan, 063210, China
| | - Zhenbang Yang
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China
| | - Zhi Zhang
- Affiliated Tangshan Gongren Hospital, North China University of Science and Technology, Tangshan, 063000, China
| | - Xuemei Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China
- College of Life Science, North China University of Science and Technology, Tangshan, 063210, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, Tangshan, 063210, China
| |
Collapse
|
46
|
Yun WG, Kim D, Han Y, Kwon W, Lee SG, Jang JY, Park D. Multiomic quantification of the KRAS mutation dosage improves the preoperative prediction of survival and recurrence in patients with pancreatic ductal adenocarcinoma. Exp Mol Med 2025; 57:193-203. [PMID: 39779977 PMCID: PMC11799340 DOI: 10.1038/s12276-024-01382-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 08/29/2024] [Accepted: 10/25/2024] [Indexed: 01/11/2025] Open
Abstract
Most cancer mutation profiling studies are laboratory-based and lack direct clinical application. For clinical use, it is necessary to focus on key genes and integrate them with relevant clinical variables. We aimed to evaluate the prognostic value of the dosage of the KRAS G12 mutation, a key pancreatic ductal adenocarcinoma (PDAC) variant and to investigate the biological mechanism of the prognosis associated with the dosage of the KRAS G12 mutation. In this retrospective cohort study, we analyzed 193 surgically treated patients with PDAC between 2009 and 2016. RNA, whole-exome, and KRAS-targeted sequencing data were used to estimate the dosage of the KRAS G12 mutant. Our prognostic scoring system included the mutation dosage from targeted sequencing ( > 0.195, 1 point), maximal tumor diameter at preoperative imaging ( > 20 mm, 1 point), and carbohydrate antigen 19-9 levels ( > 150 U/mL, 1 point). The KRAS mutation dosage exhibited comparable performance with clinical variables for survival prediction. High KRAS mutation dosages activated the cell cycle, leading to high mutation rates and poor prognosis. According to prognostic scoring systems that integrate mutation dosage with clinical factors, patients with 0 points had superior median overall survival of 97.0 months and 1-year, 3-year, and 5-year overall survival rates of 95.8%, 70.8%, and 66.4%, respectively. In contrast, patients with 3 points had worse median overall survival of only 16.0 months and 1-year, 3-year, and 5-year overall survival rates of 65.2%, 8.7%, and 8.7%, respectively. The incorporation of the KRAS G12 mutation dosage variable into prognostic scoring systems can improve clinical variable-based survival prediction, highlighting the feasibility of an integrated scoring system with clinical significance.
Collapse
Affiliation(s)
- Won-Gun Yun
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Daeun Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
- Ajou Energy Science Research Center, Ajou University, Suwon, South Korea
| | - Youngmin Han
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Wooil Kwon
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Seong-Geun Lee
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.
| | - Daechan Park
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea.
- Advanced College of Bio-convergence Engineering, Ajou University, Suwon, South Korea.
| |
Collapse
|
47
|
Zhang XJ, Lin FF, Wen YQ, Guan KP. Improving molecular subtypes and prognosis of pancreatic cancer through multi group analysis and machine learning. Discov Oncol 2025; 16:96. [PMID: 39873820 PMCID: PMC11775367 DOI: 10.1007/s12672-025-01841-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/20/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Pancreatic cancer (PAC) has a complex tumor immune microenvironment, and currently, there is a lack of accurate personalized treatment. Establishing a novel consensus machine learning driven signature (CMLS) that offers a unique predictive model and possible treatment targets for this condition was the goal of this study. METHODS This study integrated multiple omics data of PAC patients, applied ten clustering techniques and ten machine learning approaches to construct molecular subtypes for PAC, and created a new CMLS. RESULTS Using multi-omics clustering, we discovered two cancer subtypes (CSs) associated with prognosis, among which CS1 exhibited poor prognostic outcomes. Subsequently, 13 central genes were identified through screening, constituting CMLS with a significant prognostic ability. The low CMLS group had a better prognosis and was more likely to possess a "hot" tumor phenotype. The prognosis for the high CMLS group was dismal. Still, the tumor mutation burden (TMB) and tumor neoantigen burden (TNB) levels in this group of patients were higher than in the low CMLS group, which were more favorable for immune therapy response. CONCLUSION This study emphasizes that CMLS provides a beneficial instrument for early prediction of patient prognosis and screening of probable patients appropriate for immunotherapy and has broad implications for clinical practice.
Collapse
Affiliation(s)
- Xue-Jian Zhang
- Department of Laboratory, the Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Fang-Fang Lin
- Department of Laboratory, the Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Ya-Qing Wen
- Department of Laboratory, the Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Kun-Ping Guan
- Department of Laboratory, the Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China.
| |
Collapse
|
48
|
Wu Q, Xiao Q, Tang X, Li L, Song D, Zhou Y, Li B, Ren G, Luo F. DAMPs prognostic signature predicts tumor immunotherapy, and identifies immunosuppressive mechanism of pannexin 1 channels in pancreatic ductal adenocarcinoma. Front Immunol 2025; 15:1516457. [PMID: 39882247 PMCID: PMC11775746 DOI: 10.3389/fimmu.2024.1516457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 12/13/2024] [Indexed: 01/31/2025] Open
Abstract
Background Damage-associated molecular patterns (DAMPs) induced by immunogenic cell death (ICD) may be useful for the immunotherapy to patients undergoing pancreatic ductal adenocarcinoma (PDAC). The aim of this study is to predict the prognosis and immunotherapy responsiveness of PDAC patients using DAMPs-related genes. Methods K-means analysis was used to identify the DAMPs-related subtypes of 175 PDAC cases. The significance of gene mutation and immune status in different subtypes was detected. LASSO regression was used to construct a DAMPs-related prognostic signature to predict the immunotherapy responsiveness of PDAC. Subsequently, in vivo and in vitro experiments and Bulk-RNA seq were used to verify the effect of hub gene pannexin 1 (PANX1) on PDAC. Results Two subtypes were clustered based on the expression levels of DAMPs genes from 175 PDAC patients. Besides, the prognosis and immune landscape in up-regulated DAMPs expression subtypes was poor. In addition, we constructed a DAMPs-related prognostic signature that correlated with immune cell infiltration and predicted immunotherapy or chemotherapy responsiveness of patients with PDAC. Mechanically, through Bulk-RNA sequencing and experiments, we found that PANX1 promoted tumor progression and immune regulation via the ATP release to active NOD1/NFκB signaling pathway in PDAC. Conclusion Our in silico analyses established a classification system based on ICD-related DAMPs genes in PDAC, and constructed a DAMPs-related prognostic model to predict the efficacy of immunotherapy. This study will provide a new perspective for targeting the DAMPs-related molecule PANX1 in the treatment of PDAC.
Collapse
Affiliation(s)
- Qianxue Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qian Xiao
- Department of Breast and Thyroid Surgery, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Tang
- Department of Respiratory and Critical Care Medicine, The First People’s Hospital of Chongqing Liang Jiang New Area, Chongqing, China
| | - Liuying Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Daqiang Song
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Benhua Li
- Department of Clinical Laboratory, The Second People’ s Hospital of Liangshan yi Autonomous Prefecture, Xichang, China
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fang Luo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
49
|
Luo Z, Lin C, Yu C, Yuan C, Wu W, Xu X, Sun R, Jia Y, Wang Y, Shen J, Wang D, Wang S, Jiang H, Jiang B, Yang X, Xie C. Targeted Degradation of SOS1 Exhibits Potent Anticancer Activity and Overcomes Resistance in KRAS-Mutant Tumors and BCR-ABL-Positive Leukemia. Cancer Res 2025; 85:101-117. [PMID: 39437162 DOI: 10.1158/0008-5472.can-24-1093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/24/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
Son of sevenless homolog 1 (SOS1) is an essential guanine nucleotide exchange factor for RAS that also plays a critical role in the activation of the small GTPase RAC mediated by BCR-ABL in leukemogenesis. Despite this, small-molecule inhibitors targeting SOS1 have shown limited efficacy in clinical trials for KRAS-mutant cancers, and their potential as a therapeutic approach for chronic myeloid leukemia (CML) remains largely unexplored. In this study, we developed a potent SOS1 proteolysis targeting chimera (PROTAC) SIAIS562055, which was designed by connecting a CRBN ligand to an analog of the SOS1 inhibitor BI-3406. SIAIS562055 exhibited sustained degradation of SOS1 and inhibition of downstream ERK pathways, resulting in superior antiproliferative activity compared with small-molecule inhibitors. SIAIS562055 also potentiated the activity of both KRAS inhibitors in KRAS-mutant cancers and ABL inhibitors in BCR-ABL-positive CML. In KRAS-mutant xenografts, SIAIS562055 displayed promising antitumor potency as a monotherapy and enhanced ERK inhibition and tumor regression when combined with KRAS inhibitors, overcoming acquired resistance. In CML cells, SIAIS562055 promoted the active uptake of BCR-ABL inhibitors by upregulating the carnitine/organic cation transporter SLC22A4. SIAIS562055 and BCR-ABL inhibitors synergistically enhanced inhibition of ABL phosphorylation and downstream signaling, demonstrating robust antitumor activities in both mouse xenografts and primary samples from patients with CML. In summary, this study suggests that PROTAC-mediated SOS1 degradation represents an effective therapeutic strategy for treating not only KRAS-mutant cancers but also BCR-ABL-harboring leukemia. Significance: The PROTAC SIAIS562055 sustainably degrades SOS1 and inhibits downstream ERK signaling, showing strong antiproliferative activity and synergistic effects with KRAS inhibitors in KRAS-mutant cancers and BCR-ABL inhibitors in chronic myeloid leukemia.
Collapse
MESH Headings
- Humans
- SOS1 Protein/genetics
- SOS1 Protein/metabolism
- SOS1 Protein/antagonists & inhibitors
- Animals
- Mice
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Proto-Oncogene Proteins p21(ras)/genetics
- Proto-Oncogene Proteins p21(ras)/metabolism
- Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Xenograft Model Antitumor Assays
- Drug Resistance, Neoplasm/drug effects
- Proteolysis/drug effects
- Mutation
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Mice, Nude
- Female
Collapse
Affiliation(s)
- Ziwei Luo
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Chencen Lin
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | | | - Changxian Yuan
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | | | - Xiaowei Xu
- Department of Hematology, Shanghai Jiao Tong University School of Medicine Affiliated Shanghai General Hospital, Shanghai, China
| | - Renhong Sun
- Gluetacs Therapeutics (Shanghai) Co., Ltd., Shanghai, China
| | - Yan Jia
- Lingang Laboratory, Shanghai, China
| | - Yafang Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Jie Shen
- Department of Pharmacy, The SATCM Third Grade Laboratory of Traditional Chinese Medicine Preparations, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | | | - Sinan Wang
- School of Biomedical Engineering & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, China
| | - Hualiang Jiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
- Drug Discovery and Development Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Biao Jiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Xiaobao Yang
- Gluetacs Therapeutics (Shanghai) Co., Ltd., Shanghai, China
| | - Chengying Xie
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
- Lingang Laboratory, Shanghai, China
| |
Collapse
|
50
|
Nagarajan Y, Chandrasekaran N, Deepa Parvathi V. Functionalized Nanomaterials In Pancreatic Cancer Theranostics And Molecular Imaging. ChemistryOpen 2025; 14:e202400232. [PMID: 39434498 PMCID: PMC11726697 DOI: 10.1002/open.202400232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/13/2024] [Indexed: 10/23/2024] Open
Abstract
Pancreatic cancer (PC) is one of the most fatal malignancies in the world. This lethality persists due to lack of effective and efficient treatment strategies. Pancreatic ductal adenocarcinoma (PDAC) is an aggressive epithelial malignancy which has a high incidence rate and contributes to overall cancer fatalities. As of 2022, pancreatic cancer contributes to about 3 % of all cancers globally. Over the years, research has characterised germline predisposition, the origin cell, precursor lesions, genetic alterations, structural alterations, transcriptional changes, tumour heterogeneity, metastatic progression, and the tumour microenvironment, which has improved the understanding of PDAC carcinogenesis. By using molecular-based target therapies, these fundamental advancements support primary prevention, screening, early detection, and treatment. The focus of this review is the use of targeted nanoparticles as an alternative to conventional pancreatic cancer treatment due to the various side effects of the latter. The principles of nanoparticle based cancer therapy is efficient targeting of tumour cells via enhanced permeability and retention (EPR) effects and decrease the chemotherapy side effects due to their non-specificity. To increase the efficiency of existing therapies and modify target nanoparticles, several molecular markers of pancreatic cancer cells have been identified. Thus pancreatic cancer cells can be detected using appropriately functionalized nanoparticles with specific signalling molecules. Once cancer has been identified, these nanoparticles can kill the tumour by inducing hyperthermia, medication delivery, immunotherapy or gene therapy. As potent co-delivery methods for adjuvants and tumor-associated antigens; nanoparticles (NPs) have demonstrated significant promise as delivery vehicles in cancer therapy. This ensures the precise internalization of the functionalized nanoparticle and thus also activates the immune system effectively against tumor cells. This review also discusses the immunological factors behind the uptake of functionalized nanoparticles in cancer therapies. Theranostics, which combine imaging and therapeutic chemicals in a single nanocarrier, are the next generation of medicines. Pancreatic cancer treatment may be revolutionised by the development of a tailored nanocarrier with diagnostic, therapeutic, and imaging capabilities. It is extremely difficult to incorporate various therapeutic modalities into a single nanocarrier without compromising the individual functionalities. Surface modification of nanocarriers with antibodies or proteins will enable to attain multifunctionality which increases the efficiency of pancreatic cancer therapy.
Collapse
Affiliation(s)
- Yoghalakshmi Nagarajan
- Department of Biomedical SciencesFaculty of Biomedical Sciences & TechnologySri Ramachandra Institute of Higher Education and Research (SRIHER)Tamil NaduChennai600116India
| | - Natarajan Chandrasekaran
- Senior Professor & Former DirectorCentre for NanobiotechnologyVellore Institute of Technology (VIT)Vellore Campus, Tiruvalam roadTamil NaduKatpadiVellore 632014
| | - Venkatachalam Deepa Parvathi
- Department of Biomedical SciencesFaculty of Biomedical Sciences & TechnologySri Ramachandra Institute of Higher Education and Research (SRIHER)Tamil NaduChennai600116India
| |
Collapse
|