51
|
Zhao R, Yi Y, Liu H, Xu J, Chen S, Wu D, Wang L, Li F. RHOF promotes Snail1 lactylation by enhancing PKM2-mediated glycolysis to induce pancreatic cancer cell endothelial-mesenchymal transition. Cancer Metab 2024; 12:32. [PMID: 39462429 PMCID: PMC11515152 DOI: 10.1186/s40170-024-00362-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/23/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND The influence of the small Rho GTPase Rif (RHOF) on tumor growth, glycolysis, endothelial-mesenchymal transition (EMT), and the potential mechanism of RHOF in pancreatic cancer (PC) were explored. METHODS RHOF expression in PC tissues and cells was assessed by qRT-PCR and western blotting. The viability, proliferation, apoptosis, migration, and invasion of PC cells were assessed using CCK-8, colony formation, EdU, flow cytometry, scratch, and Transwell assays. The expression of EMT- and glycolysis-related proteins was determined using western blotting. The potential mechanisms of action of RHOF in PC were identified using bioinformatic analysis. The effects of RHOF were assessed in vivo using a xenograft mouse model. RESULTS PC cell proliferation, migration, and invasion are accelerated by RHOF overexpression, which inhibited apoptosis. RHOF overexpression promoted EMT and glycolysis as evidenced by a decrease in E-cadherin expression and an increase in N-cadherin, Vimentin, HK2, PKM2, and LDHA expression. Bioinformatic analysis indicated that RHOF activated EMT, glycolysis, and Myc targets and that c-Myc could bind to the PKM2 promoter. RHOF overexpression promotes the lactylation and nuclear translocation of Snail1. Silencing Snail1 reversed the promoting effects of RHOF and lactate on cell migration, invasion, and EMT. Moreover, in vivo tumor growth and EMT were inhibited by RHOF silencing. CONCLUSION RHOF plays an oncogenic role in PC. c-Myc is upregulated by RHOF and promotes PKM2 transcription. PKM2 further induces glycolysis, and the lactate produced by glycolysis causes the lactylation of Snail1, ultimately promoting EMT.
Collapse
Affiliation(s)
- Rui Zhao
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Yanmin Yi
- Department of Pancreatic Surgery, General Surgery, Qi Lu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, Shandong, 250012, China
| | - Han Liu
- Department of Pancreatic Surgery, General Surgery, Qi Lu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, Shandong, 250012, China
| | - Jianwei Xu
- Department of Pancreatic Surgery, General Surgery, Qi Lu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, Shandong, 250012, China
| | - Shuhai Chen
- Department of Pancreatic Surgery, General Surgery, Qi Lu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, Shandong, 250012, China
| | - Dong Wu
- Department of Pancreatic Surgery, General Surgery, Qi Lu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, Shandong, 250012, China
| | - Lei Wang
- Department of Pancreatic Surgery, General Surgery, Qi Lu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, Shandong, 250012, China.
| | - Feng Li
- Department of Pancreatic Surgery, General Surgery, Qi Lu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhuaxi Road, Jinan, Shandong, 250012, China.
| |
Collapse
|
52
|
Zhang D, Cui F, Zheng K, Li W, Liu Y, Wu C, Peng L, Yang Z, Chen Q, Xia C, Li S, Jin Z, Xu X, Jin G, Li Z, Huang H. Single-cell RNA sequencing reveals the process of CA19-9 production and dynamics of the immune microenvironment between CA19-9 (+) and CA19-9 (-) PDAC. Chin Med J (Engl) 2024; 137:2415-2428. [PMID: 38816396 PMCID: PMC11479433 DOI: 10.1097/cm9.0000000000003130] [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: 10/08/2023] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the main types of malignant tumor of the digestive system, and patient prognosis is affected by difficulties in early diagnosis, poor treatment response, and a high postoperative recurrence rate. Carbohydrate antigen 19-9 (CA19-9) has been widely used as a biomarker for the diagnosis and postoperative follow-up of PDAC patients. Nevertheless, the production mechanism and potential role of CA19-9 in PDAC progression have not yet been elucidated. METHODS We performed single-cell RNA sequencing on six samples pathologically diagnosed as PDAC (three CA19-9-positive and three CA19-9-negative PDAC samples) and two paracarcinoma samples. We also downloaded and integrated PDAC samples (each from three CA19-9-positive and CA19-9-negative patients) from an online database. The dynamics of the proportion and potential function of each cell type were verified through immunofluorescence. Moreover, we built an in vitro coculture cellular model to confirm the potential function of CA19-9. RESULTS Three subtypes of cancer cells with a high ability to produce CA19-9 were identified by the markers TOP2A , AQP5 , and MUC5AC . CA19-9 production bypass was discovered on antigen-presenting cancer-associated fibroblasts (apCAFs). Importantly, the proportion of immature ficolin-1 positive (FCN1+) macrophages was high in the CA19-9-negative group, and the proportion of mature M2-like macrophages was high in the CA19-9-positive group. High proportions of these two macrophage subtypes were associated with an unfavourable clinical prognosis. Further experiments indicated that CA19-9 could facilitate the transformation of M0 macrophages into M2 macrophages in the tumor microenvironment. CONCLUSIONS Our study described CA19-9 production at single-cell resolution and the dynamics of the immune atlas in CA19-9-positive and CA19-9-negative PDAC. CA19-9 could promote M2 polarization of macrophage in the pancreatic tumor microenvironment.
Collapse
Affiliation(s)
- Deyu Zhang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Fang Cui
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Kailian Zheng
- Department of Pancreatic Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Wanshun Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yue Liu
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Chang Wu
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Lisi Peng
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Zhenghui Yang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Qianqian Chen
- College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350000, China
| | - Chuanchao Xia
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Shiyu Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Zhendong Jin
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Xiaojiang Xu
- Department of Pathology and Laboratory Medicine, Tulane University, New Orleans, LA 70115 USA
- School of Medicine, Tulane University, New Orleans, LA 70115 USA
| | - Gang Jin
- Department of Pancreatic Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Zhaoshen Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Haojie Huang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
53
|
Wu J, Zhang Y, Wang H, Guo W, Li C, Yu Y, Liu H, Li F, Wang L, Xu J. Evaluating the benefits of adjuvant chemotherapy in patients with pancreatic cancer undergoing radical pancreatectomy after neoadjuvant therapy-a systematic review and meta-analysis. Front Oncol 2024; 14:1429386. [PMID: 39484040 PMCID: PMC11524795 DOI: 10.3389/fonc.2024.1429386] [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: 05/08/2024] [Accepted: 09/16/2024] [Indexed: 11/03/2024] Open
Abstract
Background More and more patients with pancreatic cancer (PC) received neoadjuvant therapy (NAT) and then underwent radical pancreatectomy. However, the benefit of adjuvant chemotherapy (AC) for these patients is still controversial. This study is designed to determine the benefits of postoperative AC for patients with PC undergoing NAT and radical resection. Methods We conducted a comprehensive search of the PubMed, Embase, Web of Science, and Cochrane Library databases, covering the period from their inception until 10 September 2023. Our analysis focused on the assessment of overall survival (OS) and recurrence-free survival (RFS) through meta-analysis. The fixed-effects model and the random-effects model were used to process the data. Hazard ratios (HRs) and 95% confidence intervals (95% CIs) were employed to determine the necessary of administering AC for patients with PC who have undergone NAT and radical resection. We retrieved 3,063 search results, of which 3,048 were excluded because of duplication or after applying our inclusion and exclusion criteria. Results A total of 15 studies with 21,113 patients (7,794 patients in the AC group and 13,319 in the non-AC group) were included, all of which reported OS, and three studies reported disease-free survival (DFS)/tumor-specific survival (CSS)/RFS. The final results showed that AC significantly improved OS and DFS/CSS/RFS in patients with PC who underwent pancreatectomy after NAT [OS: HR = 0.80, 95% CI (0.75∼0.86), P < 0.00001, I2 = 48%; DFS/CSS/RFS: HR = 0.53, 95% CI (0.41~0.69), P < 0.00001, I2 = 0%]. Furthermore, we performed subgroup analyses and demonstrated that AC provided a significant survival benefit for patients with PC after NAT and resection regardless of the tumor size [<2-cm subgroup: HR = 0.72, 95% CI (0.5∼0.94), P = 0.01; ≥2-cm subgroup: HR = 0.79, 95% CI (0.65∼0.96), P = 0.02] and the margin status [R0 subgroup: HR = 0.83, 95% CI (0.77∼0.88), P < 0.00001; R2 subgroup: HR = 0.75, 95% CI (0.61∼0.92), P = 0.007]. AC also benefited the patients with a stage N0 [HR = 0.79, 95% CI (0.74~0.84), P < 0.00001], N1 [HR = 0.78, 95% CI (0.72∼0.85), P < 0.00001], or poorly/undifferentiated tumor [HR = 0.76, 95% CI (0.66∼0.87), P < 0.0001] in survival but not in patients with a stage N2 [HR = 0.69, 95% CI (0.43∼1.09), P = 0.11] or well/moderately differentiated tumor [HR = 0.97, 95% CI (0.66∼1.42), P = 0.87]. Conclusions Although AC showed survival benefit for patients with PC undergoing radical pancreatectomy after NAT, we still need to consider the lymph node stage and the degree of differentiation of the tumor when we gave AC to a patient. High-quality prospective randomized controlled studies are required to well disclose the value of AC in patients with PC undergoing radical pancreatectomy after NAT. Systematic review registration https://www.crd.york.ac.uk/prospero/ PROSPERO, identifier CRD42023461365.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Lei Wang
- Department of Pancreatic Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Jianwei Xu
- *Correspondence: Jianwei Xu, ; Lei Wang,
| |
Collapse
|
54
|
Zhu Q, Zeng S, Yang J, Zhuo J, Wang P, Wen S, Fang C. Plectin-1-targeted recognition for enhancing comprehensive therapy in pancreatic ductal adenocarcinoma. NANOSCALE 2024; 16:18584-18596. [PMID: 39291372 DOI: 10.1039/d4nr01587h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses a formidable challenge due to its aggressive nature and poor prognosis. Gemcitabine (Gem), a primary therapeutic option, functions by inhibiting DNA synthesis and promoting apoptosis, thereby impeding the progression of PDAC. However, Gem is hindered by suboptimal pharmacokinetics and efficacy. In response to these challenges, we have developed a nanoparticle (NP) designed for specific recognition of plectin-1 in PDAC cell membranes. The NPs encapsulate Gem while demonstrating pH-responsive drug release characteristics in the acidic tumor microenvironment. This targeted approach enhances local drug delivery while alleviating concerns about systemic toxicity. Furthermore, the NPs are enriched with indocyanine green (ICG), renowned for its strong photothermal effects, thereby further enhancing therapeutic outcomes. This study presents an innovative therapeutic strategy for PDAC based on a plectin-1-targeted recognition delivery approach. The approach is applied to enhance chemotherapy, combined with photothermal therapy (PTT), inducing apoptosis in PDAC cell lines and improving the pharmacokinetics of Gem. In conclusion, the delivery strategy based on plectin-1-targeted recognition shows promising preclinical prospects for enhancing therapeutic efficacy in PDAC, offering valuable insights for future clinical applications.
Collapse
Affiliation(s)
- Qing Zhu
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Silue Zeng
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Junying Yang
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Jiaming Zhuo
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Peifeng Wang
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Sai Wen
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| | - Chihua Fang
- Department of Hepatobiliary Surgery I, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou, 510280, China
| |
Collapse
|
55
|
Fields BC, Tzeng CWD. Locally Advanced Pancreas Cancer, Is There a Role for Surgery? Surg Clin North Am 2024; 104:1017-1030. [PMID: 39237161 PMCID: PMC11748233 DOI: 10.1016/j.suc.2024.03.004] [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] [Indexed: 09/07/2024]
Abstract
Locally advanced pancreatic cancer (LAPC) represents a unique clinical scenario in which the tumor is considered localized but unresectable due to anatomic factors. Despite a consensus against upfront surgery, no standard approach to induction therapy exists for patients with LAPC. Extended systemic therapy has shown promise in establishing tumor response and remains the standard of care. While associated with improved local control, the timing and role of radiation therapy remain in question. Following adequate response to induction chemotherapy, a safe attempt at margin-negative resection can be considered. Special attention should be given to required vascular skeletonization and/or resection with reconstruction.
Collapse
Affiliation(s)
- Brittany C Fields
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX 77030, USA
| | - Ching-Wei D Tzeng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX 77030, USA.
| |
Collapse
|
56
|
Askari N, Hadizadeh M, Sina M, Parvizpour S, Mousavi SZ, Shamsir MS. Investigating the function and targeting of MET protein as an oncogene kinase in pancreatic ductal adenocarcinoma: A microarray data integration. BIOIMPACTS : BI 2024; 15:30187. [PMID: 40161938 PMCID: PMC11954745 DOI: 10.34172/bi.30187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 05/06/2024] [Accepted: 08/03/2024] [Indexed: 04/02/2025]
Abstract
Introduction Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease with a poor prognosis. Kinase proteins are essential regulators of cellular processes and potential targets for drug development. Methods Integration of multiple microarray datasets was screened to find differentially expressed kinases (DE-Kinases) across adjacent normal and tumor tissue samples in PDAC. The most effective kinase for drug design and docking in this study was selected by investigating biological mechanisms and survival analyses. Forty phytochemicals were extracted from the yellow sweet clover, Melilotus officinalis (Linn.) Pall, and were then subjected to in silico screening and molecular docking studies against a specific potent kinase. Results MET, PAK3, and PDK4 were identified as the DE-Kinases. After examining the pathways and biological processes, up-regulated MET had the most significant survival analysis and became our primary kinase for drug design and docking in this study. Four of the extracted phytocompounds of Melilotus officinalis (Linn.) Pall that exhibited high binding affinities with MET and were selected for toxicity analysis. Finally, the stability and mobility of the two nontoxic compounds that passed the toxicity test (dicumarol PubChem CID: 54676038 and melilotigenin PubChem CID: 14059499) were studied by molecular dynamics simulation. Conclusion This study's results identified two phytochemicals in yellow sweet clover that could be used to develop an anticancer drug, but experimental evaluation is necessary to confirm their efficacy.
Collapse
Affiliation(s)
- Nahid Askari
- Department of Biotechnology, Institute of Sciences and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Morteza Hadizadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Sina
- A. Nocivelli Institute for Molecular Medicine, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Sepideh Parvizpour
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyedeh Zahra Mousavi
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohd Shahir Shamsir
- Bioinformatics Research Group (BIRG), Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, Skudai, Johor, Malaysia
| |
Collapse
|
57
|
Ou Z, Yan A, Zhu W. The establishment and validation of a clinical prediction model for postoperative biliary fistula after pancreaticoduodenectomy. Gland Surg 2024; 13:1605-1618. [PMID: 39421048 PMCID: PMC11480877 DOI: 10.21037/gs-24-174] [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: 05/17/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024]
Abstract
Background At present, pancreaticoduodenectomy (PD) is a classic surgical treatment for benign and malignant tumors around ampulla. The operation is complicated and postoperative complications are frequent. Biliary fistula is the most common anastomotic fistula after pancreatic fistula. Our objective is to analyze the risk factors for biliary fistula after PD and to construct a nomogram to predict biliary fistula after PD. Methods The clinical data of a total of 196 patients who underwent PD from March 2014 to March 2024 in Yueyang Hospital Affiliated to Hunan Normal University and The Third Xiangya Hospital of Central South University were retrospectively analyzed. The number of included patients was divided in the ratio of 7:3 using the random split method, with 130 patients in the training set and 66 patients in the validation set. Predictors were screened and a nomogram prediction model was constructed by one-way logistic regression analysis, Lasso regression analysis and multifactorial logistic regression analysis. The discriminative ability, consistency and clinical effectiveness of the models were assessed by area under the curve (AUC) of the working characteristics of the subjects, calibration curve and decision curve analysis (DCA). Results The results of multifactorial logistic regression analysis showed that diabetes, low albumin, postoperative gastroparesis, abdominal bleeding, abdominal infection, and postoperative pancreatic fistula were the independent risk factors for biliary fistula after PD (P<0.05). The AUC of the column-line graph prediction model constructed from the above factors was 0.807 [95% confidence interval (CI): 0.652-0.962] in the training set and 0.782 (95% CI: 0.517-1.000) in the validation set, suggesting that the diagnostic efficacy of the model was better, and the calibration curves plotted in the training and validation sets were closer to the standard curves, suggesting that the model consistency was better. The plotted DCA curves also indicated a significant positive net gain. Conclusions The nomogram prediction model constructed by diabetes, albumin, postoperative gastroparesis, abdominal bleeding, abdominal infection, and postoperative pancreatic fistula can well identify high-risk patients with postoperative biliary fistula (POBF) in PD, which has a good clinical application value.
Collapse
Affiliation(s)
- Zhengrong Ou
- Department of General Surgery, Ward Two, Yueyang Hospital Affiliated to Hunan Normal University, Yueyang, China
| | - An Yan
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Weidong Zhu
- Department of General Surgery, Ward Two, Yueyang Hospital Affiliated to Hunan Normal University, Yueyang, China
| |
Collapse
|
58
|
Szydlak R. Synergistic Inhibition of Pancreatic Cancer Cell Growth and Migration by Gemcitabine and Withaferin A. Biomolecules 2024; 14:1178. [PMID: 39334944 PMCID: PMC11430445 DOI: 10.3390/biom14091178] [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: 08/18/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Pancreatic cancer remains one of the most lethal malignancies due to its aggressive nature and resistance to conventional therapies. This study investigates the anti-proliferative, pro-apoptotic, and anti-migratory effects of Gemcitabine (GC) and Withaferin A (WFA) on pancreatic cancer cell lines PANC-1 and Hs766t. The MTS assay revealed that both compounds effectively inhibit cell proliferation, with WFA showing a stronger effect in Hs766t cells. Flow cytometry analysis demonstrated that GC and WFA, particularly in combination, significantly induce apoptosis in both cell lines. Migration assays confirmed the potent inhibition of cell migration by both compounds, with the combination treatment being the most effective. Furthermore, actin cytoskeleton analysis indicated substantial changes in cell morphology and stiffness, suggesting that GC and WFA disrupt the structural integrity of cancer cells. Additionally, the study highlights a ROS-mediated mechanism underlying the effects of GC and WFA, as evidenced by increased ROS levels following treatment, which were attenuated by N-acetylcysteine. Importantly, NF-κB activity was significantly modulated, with WFA reducing NF-κB activation induced by GC, potentially contributing to the synergistic pro-apoptotic effect of the combination. These findings suggest that the combination of GC and WFA may offer a synergistic therapeutic approach for treating pancreatic cancer by targeting multiple aspects of tumor cell behavior.
Collapse
Affiliation(s)
- Renata Szydlak
- Department of Bioinformatics and Telemedicine, Faculty of Medicine, Jagiellonian University Medical College, Medyczna 7, 30-688 Krakow, Poland
| |
Collapse
|
59
|
Shapoval O, Patsula V, Větvička D, Engstová H, Oleksa V, Kabešová M, Vasylyshyn T, Poučková P, Horák D. Temoporfin-Conjugated PEGylated Poly( N, N-dimethylacrylamide)-Coated Upconversion Colloid for NIR-Induced Photodynamic Therapy of Pancreatic Cancer. Biomacromolecules 2024; 25:5771-5785. [PMID: 38888278 PMCID: PMC11388470 DOI: 10.1021/acs.biomac.4c00317] [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: 03/07/2024] [Revised: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024]
Abstract
Photodynamic therapy (PDT) has the potential to cure pancreatic cancer with minimal side effects. Visible wavelengths are primarily used to activate hydrophobic photosensitizers, but in clinical practice, these wavelengths do not sufficiently penetrate deeper localized tumor cells. In this work, NaYF4:Yb3+,Er3+,Fe2+ upconversion nanoparticles (UCNPs) were coated with polymer and labeled with meta-tetra(hydroxyphenyl)chlorin (mTHPC; temoporfin) to enable near-infrared light (NIR)-triggered PDT of pancreatic cancer. The coating consisted of alendronate-terminated poly[N,N-dimethylacrylamide-co-2-aminoethylacrylamide]-graft-poly(ethylene glycol) [P(DMA-AEM)-PEG-Ale] to ensure the chemical and colloidal stability of the particles in aqueous physiological fluids, thereby also improving the therapeutic efficacy. The designed particles were well tolerated by the human pancreatic adenocarcinoma cell lines CAPAN-2, PANC-1, and PA-TU-8902. After intratumoral injection of mTHPC-conjugated polymer-coated UCNPs and subsequent exposure to 980 nm NIR light, excellent PDT efficacy was achieved in tumor-bearing mice.
Collapse
Affiliation(s)
- Oleksandr Shapoval
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského Nám. 2, 162
00 Prague 6, Czech
Republic
| | - Vitalii Patsula
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského Nám. 2, 162
00 Prague 6, Czech
Republic
| | - David Větvička
- First
Faculty of Medicine, Charles University, Salmovská 1, 120 00 Prague 2, Czech Republic
| | - Hana Engstová
- Institute
of Physiology, Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Viktoriia Oleksa
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského Nám. 2, 162
00 Prague 6, Czech
Republic
| | - Martina Kabešová
- First
Faculty of Medicine, Charles University, Salmovská 1, 120 00 Prague 2, Czech Republic
| | - Taras Vasylyshyn
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského Nám. 2, 162
00 Prague 6, Czech
Republic
| | - Pavla Poučková
- First
Faculty of Medicine, Charles University, Salmovská 1, 120 00 Prague 2, Czech Republic
| | - Daniel Horák
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského Nám. 2, 162
00 Prague 6, Czech
Republic
| |
Collapse
|
60
|
Lu PW, Lyu HG, Prakash LR, Chiang YJS, Maxwell JE, Snyder RA, Kim MP, Tzeng CWD, Katz MHG, Ikoma N. Effect of surgical approach on early return to intended oncologic therapy after resection for pancreatic ductal adenocarcinoma. Surg Endosc 2024; 38:4986-4995. [PMID: 38987482 DOI: 10.1007/s00464-024-11022-3] [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: 03/08/2024] [Accepted: 06/30/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Although robotic pancreatectomy may facilitate an earlier functional recovery, the impact of a robotic pancreatectomy program during its early experience on the timing of return to intended oncologic therapy (RIOT) after surgery is unknown. METHODS In this retrospective cohort study, we used propensity score matching with a 1:2 ratio to compare patients who underwent robotic or open surgery (distal pancreatectomy or pancreatoduodenectomy) for pancreatic ductal adenocarcinoma (PDAC) during the first 3 years of our robotic pancreatectomy experience (January 2018-December 2021). Generalized estimating equations modeling was used to evaluate the effect of surgical approach on early RIOT, defined as adjuvant chemotherapy initiation within 8 weeks after surgery, and late RIOT, defined as initiation within 12 weeks after surgery. RESULTS The matched cohort included 26 patients who underwent robotic pancreatectomy and 52 patients who underwent open pancreatectomy. Rates of receipt of adjuvant chemotherapy were 96.2% and 78.9%, respectively. Rate of early RIOT in the robotic group (73.1% was higher than that in the open group (44.2%; P = 0.018). In multivariable analysis, a robotic approach was associated with early RIOT (odds ratio, 3.54; 95% confidence interval 1.08-11.62; P = 0.038). Surgical approach did not impact late RIOT (odds ratio, 3.21; 95% confidence interval 0.71-14.38; P = 0.128). CONCLUSIONS Compared with open pancreatectomy, robotic pancreatectomy did not delay RIOT. In fact, odds of early RIOT were increased, which supports the oncological safety of our robotic pancreatectomy program during its implementation.
Collapse
Affiliation(s)
- Pamela W Lu
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Heather G Lyu
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Laura R Prakash
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Yi-Ju Sabrina Chiang
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Jessica E Maxwell
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Rebecca A Snyder
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Michael P Kim
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Ching-Wei D Tzeng
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Matthew H G Katz
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Naruhiko Ikoma
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| |
Collapse
|
61
|
Oguma T, Kanazawa T, Kaneko YK, Sato R, Serizawa M, Ooka A, Yamaguchi M, Ishikawa T, Kondo H. Effects of phospholipid type and particle size on lipid nanoparticle distribution in vivo and in pancreatic islets. J Control Release 2024; 373:917-928. [PMID: 39079658 DOI: 10.1016/j.jconrel.2024.07.059] [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: 03/15/2024] [Revised: 07/03/2024] [Accepted: 07/25/2024] [Indexed: 09/11/2024]
Abstract
Lipid nanoparticles (LNPs) have recently been used as nanocarriers in drug delivery systems for nucleic acid drugs. Their practical applications are currently primarily limited to the liver and specific organs. However, altering the type and composition ratio of phospholipids improves their distribution in organs other than the liver, such as the spleen and lungs. This study aimed to elucidate the effects of LNP components and particle size on in vivo distribution through systemic circulation to pancreatic islets to achieve better targeting of islets, which are a fundamental therapeutic target for diabetes. Fluorescence-labeled LNPs were prepared using three phospholipids: 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), and 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), with particle sizes of 30-160 nm (diameter) using a microfluidic device. Baffled-structured iLiNP devices with adjusted flow-rate ratios and total flow rates were used. After the intravenous administration of LNPs to C57BL/6 J mice, the distribution of each LNP type to the major organs, including the pancreas and pancreatic islets, was compared using ex vivo fluorescence imaging and observation of pancreatic tissue sections. DSPC-LNPs- and DOPE-LNPs showed the highest distribution in the spleen and liver, respectively. In contrast, the DOPC-LNPs showed the highest distribution in the pancreas and the lowest distribution in the liver and spleen. In addition, smaller particles showed better distribution throughout the pancreas. The most significant LNP distribution in the islets was observed for DOPC-LNPs with a particle size of 160 nm. Furthermore, larger LNPs tended to be distributed in the islets, whereas smaller LNPs tended to be distributed in the exocrine glands. DOPC-LNPs were distributed in the islets at all cholesterol concentrations, with a high distribution observed at >40% cholesterol and > 3% PEG and the distribution was higher at 24 h than at 4 h. Thus, LNP composition and particle size significantly affected islet distribution characteristics, indicating that DOPC-LNPs may be a drug delivery system for effectively targeting the pancreas and islets.
Collapse
Affiliation(s)
- Takayuki Oguma
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; Center for Clinical Research, Hamamatsu University School of Medicine, 1-20-1, Handayama, Chuo-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Takanori Kanazawa
- Department of Pharmaceutical Engineering and Drug Delivery Sciences, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; Department of Clinical Pharmacology, Graduate School of Biomedical Sciences, Tokushima University, 1-78-1, Shoumachi, Tokushima 770-8505, Japan; Innovative Research Center for Drug Delivery System, Graduate School of Biomedical Sciences, Tokushima University, 1-78-1, Shoumachi, Tokushima 770-8505, Japan.
| | - Yukiko K Kaneko
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| | - Ren Sato
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Miku Serizawa
- Department of Pharmaceutical Engineering and Drug Delivery Sciences, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Akira Ooka
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Momoka Yamaguchi
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Tomohisa Ishikawa
- Department of Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Hiromu Kondo
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; Department of Pharmaceutical Engineering and Drug Delivery Sciences, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| |
Collapse
|
62
|
Chen X, Zhang H. Comprehensive exploration of immune checkpoint-related genes in the prognosis and tumor immune microenvironment of pancreatic adenocarcinoma. Clinics (Sao Paulo) 2024; 79:100481. [PMID: 39208654 PMCID: PMC11399560 DOI: 10.1016/j.clinsp.2024.100481] [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] [Received: 05/13/2024] [Revised: 07/23/2024] [Accepted: 07/28/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND To comprehensively analyze the clinical significance of Immune Checkpoint-Related Genes (ICRGs) in Pancreatic Adenocarcinoma (PAAD). METHOD PAAD tissues and normal pancreatic tissues were obtained from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases, and 283 ICRGs were integrated by the Kyoto Encyclopedia of Genes and Genomes (KEGG) and Reactome datasets. Unsupervised clustering was used to obtain potential ICRGs-based PAAD subtypes. Wilcoxon test was performed to screen Differentially Expressed ICRGs (DEICRGs), while cox regression analyses were utilized to identify prognosis-related ICRGs and clinicopathological factors, and construct the corresponding models. The Tumor Immune Microenvironment (TIME) was evaluated. Moreover, the authors performed enrichment analysis, Gene Set Enrichment Analysis (GSEA), and transcription factor regulatory networks to realize underlying mechanisms. RESULTS Three ICRGs-based PAAD subtypes were identified, and they were associated with three ESTIMATE scores, a Tumor Microenvironment (TMB) score, 14 therapeutic immune checkpoints, and infiltration levels of seven immune cells. On top of that, the authors constructed two signatures based on DEICRGs to predict the Overall Survival (OS) (Area Under the ROC Curve [AUC: 0.741∼0.778]) and Progression-Free Survival (PFS) (AUC: 0.746∼0.831) of patients. Two nomograms were established by combining clinical variables and signatures. In addition, the authors found higher infiltration of naïve B cells and CD8+ T-cells in low-risk PAAD patients, and higher infiltration of suppressive immune cells and cancer-related signaling pathways in high-risk PAAD patients. CONCLUSION The present study suggested that ICRGs were associated with TIME formation and prognosis of PAAD patients, which may serve as novel clinical biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Surgery, Suzhou Hospital of Anhui Medical University, Suzhou, PR China
| | - Herui Zhang
- Department of Gastroenterology, Suzhou Hospital of Anhui Medical University, Suzhou, PR China.
| |
Collapse
|
63
|
Shao L, Yu H, Wang M, Chen L, Ji B, Wu T, Teng X, Su M, Han X, Shi W, Hu X, Wang Z, He H, Han G, Zhang Y, Wu Q. DKK1-SE recruits AP1 to activate the target gene DKK1 thereby promoting pancreatic cancer progression. Cell Death Dis 2024; 15:566. [PMID: 39107271 PMCID: PMC11303742 DOI: 10.1038/s41419-024-06915-z] [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: 02/21/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 08/09/2024]
Abstract
Super-enhancers are a class of DNA cis-regulatory elements that can regulate cell identity, cell fate, stem cell pluripotency, and even tumorigenesis. Increasing evidence shows that epigenetic modifications play an important role in the pathogenesis of various types of cancer. However, the current research is far from enough to reveal the complex mechanism behind it. This study found a super-enhancer enriched with abnormally active histone modifications in pancreatic ductal adenocarcinoma (PDAC), called DKK1-super-enhancer (DKK1-SE). The major active component of DKK1-SE is component enhancer e1. Mechanistically, AP1 induces chromatin remodeling in component enhancer e1 and activates the transcriptional activity of DKK1. Moreover, DKK1 was closely related to the malignant clinical features of PDAC. Deletion or knockdown of DKK1-SE significantly inhibited the proliferation, colony formation, motility, migration, and invasion of PDAC cells in vitro, and these phenomena were partly mitigated upon rescuing DKK1 expression. In vivo, DKK1-SE deficiency not only inhibited tumor proliferation but also reduced the complexity of the tumor microenvironment. This study identifies that DKK1-SE drives DKK1 expression by recruiting AP1 transcription factors, exerting oncogenic effects in PDAC, and enhancing the complexity of the tumor microenvironment.
Collapse
Affiliation(s)
- Lan Shao
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, China
| | - Haoran Yu
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, China
| | - Mengyun Wang
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, China
| | - Lu Chen
- Department of Pathology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Boshu Ji
- Department of Pathology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tong Wu
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, China
| | - Xiangqi Teng
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, China
| | - Mu Su
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, China
| | - Xiao Han
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, China
| | - Weikai Shi
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, China
| | - Xin Hu
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, China
| | - Ziwen Wang
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, China
| | - Hongjuan He
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, China
| | - Guiping Han
- Department of Pathology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yan Zhang
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, China
| | - Qiong Wu
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, China.
| |
Collapse
|
64
|
Hong J, Du K, Zhang W, Chen J, Jin H, Chen Y, Jiang Y, Yu H, Weng X, Zheng S, Yu J, Cao L. 6:2 Cl-PFESA, a proposed safe alternative for PFOS, diminishes the gemcitabine effectiveness in the treatment of pancreatic cancer. JOURNAL OF HAZARDOUS MATERIALS 2024; 474:134790. [PMID: 38850938 DOI: 10.1016/j.jhazmat.2024.134790] [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: 04/09/2024] [Revised: 05/23/2024] [Accepted: 05/31/2024] [Indexed: 06/10/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC)/pancreatic cancer, is a highly aggressive malignancy with poor prognosis. Gemcitabine-based chemotherapy remains the cornerstone of PDAC treatment. Nonetheless, the development of resistance to gemcitabine among patients is a major factor contributing to unfavorable prognostic outcomes. The resistance exhibited by tumors is modulated by a constellation of factors such as genetic mutations, tumor microenvironment transforms, environmental contaminants exposure. Currently, comprehension of the relationship between environmental pollutants and tumor drug resistance remains inadequate. Our study found that PFOS/6:2 Cl-PFESA exposure increases resistance to gemcitabine in PDAC. Subsequent in vivo trials confirmed that exposure to PFOS/6:2 Cl-PFESA reduces gemcitabine's efficacy in suppressing PDAC, with the inhibition rate decreasing from 79.5 % to 56.7 %/38.7 %, respectively. Integrative multi-omics sequencing and molecular biology analyses have identified the upregulation of ribonucleotide reductase catalytic subunit M1 (RRM1) as a critical factor in gemcitabine resistance. Subsequent research has demonstrated that exposure to PFOS and 6:2 Cl-PFESA results in the upregulation of the RRM1 pathway, consequently enhancing chemotherapy resistance. Remarkably, the influence exerted by 6:2 Cl-PFESA exceeds that of PFOS. Despite 6:2 Cl-PFESA being regarded as a safer substitute for PFOS, its pronounced effect on chemotherapeutic resistance in PDAC necessitates a thorough evaluation of its potential risks related to gastrointestinal toxicity.
Collapse
Affiliation(s)
- Jiawei Hong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
| | - Keyi Du
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
| | - Weichen Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
| | - Junran Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
| | - Hangbiao Jin
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang 310032, China; Innovation Research Center of Advanced Environmental Technology, Eco-Industrial Innovation Institute ZJUT, Quzhou, Zhejiang 324400, China
| | - Yuanchen Chen
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang 310032, China; Innovation Research Center of Advanced Environmental Technology, Eco-Industrial Innovation Institute ZJUT, Quzhou, Zhejiang 324400, China
| | - Yifan Jiang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
| | - Hanxi Yu
- Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Xiaoyu Weng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
| | - Jun Yu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
| | - Linping Cao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China.
| |
Collapse
|
65
|
Zeppa L, Aguzzi C, Morelli MB, Marinelli O, Amantini C, Giangrossi M, Santoni G, Fanelli A, Luongo M, Nabissi M. In Vitro and In Vivo Effects of Melatonin-Containing Combinations in Human Pancreatic Ductal Adenocarcinoma. J Pineal Res 2024; 76:e12997. [PMID: 39076059 DOI: 10.1111/jpi.12997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 06/20/2024] [Accepted: 07/17/2024] [Indexed: 07/31/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has poor prognosis and high mortality rates. Therefore, it is necessary to identify new targets and therapeutic strategies to improve the prognosis of patients with PDAC. Integrative therapies are increasingly being used to boost the efficacy of the known anticancer therapeutic approaches. Hence, this study aimed to evaluate the effects of a novel combination of different potential anticancer molecules, melatonin (MLT), cannabidiol (CBD), and oxygen-ozone (O2/O3) to treat PDAC using in vitro and in vivo models of human PDAC. The effect of this combination was investigated in combination with gemcitabine (GEM), the most common chemotherapeutic drug used for PDAC treatment. The combination of MLT + CBD + O2/O3 was more effective than the individual treatments in inhibiting PDAC cell viability and proliferation, inducing cell death, and modulating the RAS pathway protein levels. Moreover, different combinations of treatments reduced tumor mass in the PDAC mouse model, thus promoting the effect of GEM. In conclusion, a mixture of MLT + CBD + O2/O3 could serve as a potential adjuvant therapeutic strategy for PDAC.
Collapse
Affiliation(s)
- Laura Zeppa
- School of Pharmacy, University of Camerino, Camerino, Macerata, Italy
- Integrative Therapy Discovery Lab, University of Camerino, Camerino, Macerata, Italy
| | - Cristina Aguzzi
- School of Pharmacy, University of Camerino, Camerino, Macerata, Italy
- Integrative Therapy Discovery Lab, University of Camerino, Camerino, Macerata, Italy
| | - Maria Beatrice Morelli
- School of Pharmacy, University of Camerino, Camerino, Macerata, Italy
- Integrative Therapy Discovery Lab, University of Camerino, Camerino, Macerata, Italy
| | - Oliviero Marinelli
- School of Pharmacy, University of Camerino, Camerino, Macerata, Italy
- Integrative Therapy Discovery Lab, University of Camerino, Camerino, Macerata, Italy
| | - Consuelo Amantini
- School of Bioscience and Veterinary Medicine, University of Camerino, Camerino, Macerata, Italy
| | | | - Giorgio Santoni
- School of Pharmacy, University of Camerino, Camerino, Macerata, Italy
| | - Alessandro Fanelli
- Department of Radiotherapy, Institute Ecomedica Empoli, Empoli, Florence, Italy
| | - Margherita Luongo
- "Maria Guarino" Foundation-AMOR No Profit Association, Pozzuoli, Naples, Italy
- Università degli Studi della Campania Luigi Vanvitelli Scuola di Medicina e Chirurgia, Naples, Italy
| | - Massimo Nabissi
- School of Pharmacy, University of Camerino, Camerino, Macerata, Italy
- Integrative Therapy Discovery Lab, University of Camerino, Camerino, Macerata, Italy
| |
Collapse
|
66
|
Li Y, Fan Z, Meng Y, Yang J, Li P, Liu S, Pang C, Du L, Wang Y, Zhan H. ZFHX3 methylation in peripheral blood monocytes as a potential biomarker for pancreatic cancer detection. Pancreatology 2024; 24:771-778. [PMID: 38853072 DOI: 10.1016/j.pan.2024.05.529] [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] [Received: 04/16/2024] [Revised: 05/26/2024] [Accepted: 05/31/2024] [Indexed: 06/11/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is the digestive malignancy with poor prognosis, and there is still a lack of effective diagnostic biomarkers. OBJECTIVE We aimed to explore the diagnostic efficiency of DNA methylation in peripheral blood monocytes (PBMCs) in PDAC. METHODS 850K BeadChips were used to detect genome-wide methylation of PBMCs. For the selected sites, MethylTarget assays was used for further verification. The support vector machine was used to establish the combined panel. RESULTS A total of 167 PDAC patients and 113 healthy controls were included in this study and were divided into three sets. In the discovery set, we found 4625 differentially methylated positions (DMPs) between cancer group and healthy controls. ZFHX3 (0.16 ± 0.04 vs. 0.18 ± 0.04, P = 0.001), cg01904886 (0.84 ± 0.05 vs. 0.81 ± 0.04, P = 0.02) and NUMBL (0.96 ± 0.005 vs. 0.957 ± 0.005, P = 0.04) were found to be significantly different in training set. The locus with more significant differences, namely ZFHX3, was used for further validation and to establish a combined diagnostic panel with CA19-9. In the validation set, the ROC curve indicated that the AUC value of ZFHX3 was 0.75. The AUC value of the combined model (AUC = 0.92) was higher than that of CA19-9 alone (AUC = 0.88). In patients with normal CA19-9 levels, the ZFHX3 methylation biomarker still maintained good diagnostic efficacy (AUC = 0.71). CONCLUSION Our study preliminarily suggests that ZFHX3 methylation combined with CA19-9 can improve the detection rate of PDAC. Especially in patients with normal CA19-9, ZFHX3 methylation can maintain stable diagnostic efficacy. The diagnostic value of ZFHX3 methylation still needs to be prospectively validated.
Collapse
Affiliation(s)
- Yongzheng Li
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China; Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Zhiyao Fan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China
| | - Yufan Meng
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China
| | - Jian Yang
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China
| | - Peilong Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Shujie Liu
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China
| | - Chaoyu Pang
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Yunshan Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Hanxiang Zhan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
67
|
Zhang Z, Yu G, Eresen A, Chen Z, Yu Z, Yaghmai V, Zhang Z. Dendritic cell vaccination combined with irreversible electroporation for treating pancreatic cancer-a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2024; 12:77. [PMID: 39118942 PMCID: PMC11304422 DOI: 10.21037/atm-23-1882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 02/25/2024] [Indexed: 08/10/2024]
Abstract
Background and Objective Pancreatic ductal adenocarcinoma (PDAC) is 3rd most lethal cancer in the USA leading to a median survival of six months and less than 5% 5-year overall survival (OS). As the only potentially curative treatment, surgical resection is not suitable for up to 90% of the patients with PDAC due to late diagnosis. Highly fibrotic PDAC with an immunosuppressive tumor microenvironment restricts cytotoxic T lymphocyte (CTL) infiltration and functions causing limited success with systemic therapies like dendritic cell (DC)-based immunotherapy. In this study, we investigated the potential benefits of irreversible electroporation (IRE) ablation therapy in combination with DC vaccine therapy against PDAC. Methods We performed a literature search to identify studies focused on DC vaccine therapy and IRE ablation to boost therapeutic response against PDAC indexed in PubMed, Web of Science, and Scopus until February 20th, 2023. Key Content and Findings IRE ablation destructs tumor structure while preserving extracellular matrix and blood vessels facilitating local inflammation. The studies demonstrated IRE ablation reduces tumor fibrosis and promotes CTL tumor infiltration to PDAC tumors in addition to boosting immune response in rodent models. The administration of the DC vaccine following IRE ablation synergistically enhances therapeutic response and extends OS rates compared to the use of DC vaccination or IRE alone. Moreover, the implementation of data-driven approaches further allows dynamic and longitudinal monitoring of therapeutic response and OS following IRE plus DC vaccine immunoablation. Conclusions The combination of IRE ablation and DC vaccine immunotherapy is a potent strategy to enhance the therapeutic outcomes in patients with PDAC.
Collapse
Affiliation(s)
- Zigeng Zhang
- Department of Radiological Sciences, University of California Irvine, Irvine, CA, USA
| | - Guangbo Yu
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Aydin Eresen
- Department of Radiological Sciences, University of California Irvine, Irvine, CA, USA
| | - Zhilin Chen
- Department of Human Biology and Business Administration, University of Southern California, Los Angeles, CA, USA
| | - Zeyang Yu
- Information School, University of Washington, Seattle, WA, USA
| | - Vahid Yaghmai
- Department of Radiological Sciences, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
| | - Zhuoli Zhang
- Department of Radiological Sciences, University of California Irvine, Irvine, CA, USA
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
68
|
Seiki Y, Ikezawa K, Watsuji K, Urabe M, Kai Y, Takada R, Yamai T, Mukai K, Nakabori T, Uehara H, Ishibashi M, Ohkawa K. Zinc supplementation for dysgeusia in patients with unresectable pancreatic cancer. Int J Clin Oncol 2024; 29:1173-1181. [PMID: 38724773 DOI: 10.1007/s10147-024-02544-w] [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: 11/02/2023] [Accepted: 04/30/2024] [Indexed: 07/26/2024]
Abstract
BACKGROUND Although patients with advanced pancreatic cancer (PC) often experience dysgeusia with zinc deficiency during chemotherapy, data on zinc supplementation for dysgeusia and its effects on nutritional status are scarce. We aimed to examine the efficacy of zinc supplementation in patients with advanced PC. METHODS Thirty-three patients with unresectable PC who presented with dysgeusia and zinc deficiency during chemotherapy and received zinc acetate hydrate between January 2018 and December 2022 were included. We evaluated the changes in serum zinc levels and the improvement in dysgeusia. Among the 26 patients who received zinc supplementation for 12 weeks, we also compared patient characteristics and changes in serum zinc and albumin levels between patients who showed improvement in dysgeusia (effective group) and those who did not (non-effective group). RESULTS The serum zinc level increased significantly after zinc supplementation (median: 60 µg/dL at baseline, 99.5 µg/dL at 4 weeks, 101 µg/dL at 8 weeks and 101 µg/dL at 12 weeks). The rate of improvement in dysgeusia increased over time (18.2% at 4 weeks, 33.3% at 8 weeks, and 42.4% at 12 weeks). Comparing the effective group and non-effective group revealed that while the median serum albumin level of the effective group did not change, the non-effective group showed a significant decrease from baseline to 12 weeks (3.2 g/dL to 3.0 g/dL, p = 0.03). CONCLUSION Zinc supplementation significantly increased serum zinc levels, improving dysgeusia. Zinc supplementation might also contribute to maintaining nutritional status in patients with unresectable PC.
Collapse
Affiliation(s)
- Yusuke Seiki
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Kenji Ikezawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan.
| | - Ko Watsuji
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Makiko Urabe
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Yugo Kai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Ryoji Takada
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Takuo Yamai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Kaori Mukai
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Tasuku Nakabori
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Hiroyuki Uehara
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Miki Ishibashi
- Department of Dentistry, Osaka International Cancer Institute, Osaka, Japan
| | - Kazuyoshi Ohkawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| |
Collapse
|
69
|
Song W, Hu H, Yuan Z, Yao H. A prognostic model for anoikis-related genes in pancreatic cancer. Sci Rep 2024; 14:15200. [PMID: 38956290 PMCID: PMC11220081 DOI: 10.1038/s41598-024-65981-7] [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: 04/18/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024] Open
Abstract
Anoikis, a distinct form of programmed cell death, is crucial for both organismal development and maintaining tissue equilibrium. Its role extends to the proliferation and progression of cancer cells. This study aimed to establish an anoikis-related prognostic model to predict the prognosis of pancreatic cancer (PC) patients. Gene expression data and patient clinical profiles were sourced from The Cancer Genome Atlas (TCGA-PAAD: Pancreatic Adenocarcinoma) and the International Cancer Genome Consortium (ICGC-PACA: Pancreatic Ductal Adenocarcinoma). Non-cancerous pancreatic tissue gene expression data were obtained from the Genotype-Tissue Expression (GTEx) project. The R package was used to construct anoikis-related PC prognostic models, which were later validated with the ICGC-PACA database. Survival analyses demonstrated a poorer prognosis for patients in the high-risk group, consistent across both TCGA-PAAD and ICGC-PACA datasets. A nomogram was designed as a predictive tool to estimate patient mortality. The study also analyzed tumor mutations and immune infiltration across various risk groups, uncovering notable differences in tumor mutation patterns and immune landscapes between high- and low-risk groups. In conclusion, this research successfully developed a prognostic model centered on anoikis-related genes, offering a novel tool for predicting the clinical trajectory of PC patients.
Collapse
Affiliation(s)
- Wenbin Song
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin, 300052, People's Republic of China
| | - Haiyang Hu
- Department of Cardiac Critical Care Medicine, Affiliated Hospital of Jining Medical University, Jining, 272007, People's Republic of China
| | - Zhengbo Yuan
- School of Medicine, Xiamen University, No.4221 Xiangan South Road, Xiangan District, Xiamen, 361102, People's Republic of China.
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, No.55 Zhenghai load, Siming District, Xiamen, 361001, People's Republic of China.
| | - Hao Yao
- Department of Hepatological Surgery, The Second Hospital of Tianjin Medical University, No.23 Pingjiang Road, Hexi District, Tianjin, 300211, People's Republic of China.
| |
Collapse
|
70
|
Chen Y, Xia H, Zhong X. In Vitro evaluation of the anti-pancreatic cancer activity of epimedium herb. Front Pharmacol 2024; 15:1389221. [PMID: 39011503 PMCID: PMC11246921 DOI: 10.3389/fphar.2024.1389221] [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: 02/21/2024] [Accepted: 06/13/2024] [Indexed: 07/17/2024] Open
Abstract
Introduction: Pancreatic cancer (PC) is a particularly aggressive malignancy with limited therapeutic options. The search for innovative treatments has focused on traditional Chinese medicine, specifically epimedium. This research investigates epimedium's active ingredients, potential targets, and underlying mechanisms in treating PC. Methods: High-performance liquid chromatography (HPLC) was used to quantify the active components of epimedium and HPLC-Q-TOF-MS was employed for qualitative identification. Potential targets of epimedium's active ingredients were identified using the TCMSP, ETCM, CTD, and Swiss Target Prediction databases. Potential PC-related targets were sourced from DisGeNET, GeneCards, and OMIM databases. A Venn diagram was utilized to identify overlapping PC-related and epimedium targets. Core targets and pathways were elucidated through protein-protein interaction (PPI) network analysis, Gene Ontology (GO) assessments, and Reactome pathway enrichment analyses. Molecular docking techniques investigated interactions between active compounds and these targets. The expression and prognostic implications of target genes were evaluated using GEPIA2 and the Human Protein Atlas (HPA) databases. In vitro studies assessed the impact of epimedium extract (EPE) on Panc-1 cell viability, and Western blot analysis examined the expression levels of key targets. Results: Network pharmacological indicate that epimedium econtains active components such as baohuoside I, icariin, hyperoside, and epimedin B, which have potential therapeutic effects against PC. In vitro assays confirmed that EPE significantly reduced the viability of Panc-1 cells. Western blot analysis revealed a considerable decrease in the expression of key targets in EPE-treated cells, including AKT1, EGFR, p-EGFR, JUN, BCL2, IL6, and SRC. The R-HSA-1280215: Interleukin-4 and Interleukin-13 signaling pathways involving these genes were identified as potential therapeutic targets. Discussion: Epimedium holds promise as a candidate for treating PC. The modulation of interleukin-4 and interleukin-13 signaling pathways could be a pivotal mechanism by which epimedium impedes tumor development. Further research is warranted to validate these findings and explore the clinical applicability of epimedium in PC treatment.
Collapse
Affiliation(s)
- Yangfeng Chen
- College of Horticulture, Hunan Agricultural University, Changsha, China
| | - Han Xia
- Changsha Central Hospital, Changsha, China
| | - Xiaohong Zhong
- College of Horticulture, Hunan Agricultural University, Changsha, China
| |
Collapse
|
71
|
Sun Y, Hu J, Wang R, Du X, Zhang X, E J, Zheng S, Zhou Y, Mou R, Li X, Zhang H, Xu Y, Liao Y, Jiang W, Liu L, Wang R, Zhu J, Xie R. Meaningful nomograms based on systemic immune inflammation index predicted survival in metastatic pancreatic cancer patients receiving chemotherapy. Cancer Med 2024; 13:e7453. [PMID: 38986683 PMCID: PMC11236459 DOI: 10.1002/cam4.7453] [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: 12/19/2023] [Revised: 05/15/2024] [Accepted: 06/24/2024] [Indexed: 07/12/2024] Open
Abstract
OBJECTIVE The purpose of the study is to construct meaningful nomogram models according to the independent prognostic factor for metastatic pancreatic cancer receiving chemotherapy. METHODS This study is retrospective and consecutively included 143 patients from January 2013 to June 2021. The receiver operating characteristic (ROC) curve with the area under the curve (AUC) is utilized to determine the optimal cut-off value. The Kaplan-Meier survival analysis, univariate and multivariable Cox regression analysis are exploited to identify the correlation of inflammatory biomarkers and clinicopathological features with survival. R software are run to construct nomograms based on independent risk factors to visualize survival. Nomogram model is examined using calibration curve and decision curve analysis (DCA). RESULTS The best cut-off values of 966.71, 0.257, and 2.54 for the systemic immunological inflammation index (SII), monocyte-to-lymphocyte ratio (MLR), and neutrophil-to-lymphocyte ratio (NLR) were obtained by ROC analysis. Cox proportional-hazards model revealed that baseline SII, history of drinking and metastasis sites were independent prognostic indices for survival. We established prognostic nomograms for primary endpoints of this study. The nomograms' predictive potential and clinical efficacy have been evaluated by calibration curves and DCA. CONCLUSION We constructed nomograms based on independent prognostic factors, these models have promising applications in clinical practice to assist clinicians in personalizing the management of patients.
Collapse
Affiliation(s)
- Yanan Sun
- Department of Digestive Internal MedicineHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Jiahe Hu
- Department of Digestive Internal MedicineHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Rongfang Wang
- Department of Digestive Internal MedicineHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Xinlian Du
- Department of Digestive Internal MedicineHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Xiaoling Zhang
- Department of Digestive Internal MedicineHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Jiaoting E
- Department of Digestive Internal MedicineHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Shaoyue Zheng
- Department of Digestive Internal MedicineHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Yuxin Zhou
- Department of Digestive Internal MedicineHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Ruishu Mou
- Department of Digestive Internal MedicineHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Xuedong Li
- Department of Digestive Internal MedicineHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Hanbo Zhang
- Department of Digestive Internal MedicineHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Ying Xu
- Department of Digestive Internal MedicineHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Yuan Liao
- Harbin Medical UniversityHarbinHeilongjiangChina
| | - Wenjie Jiang
- Harbin Medical UniversityHarbinHeilongjiangChina
| | - Lijia Liu
- Harbin Medical UniversityHarbinHeilongjiangChina
| | - Ruitao Wang
- Department of Internal MedicineHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| | - Jiuxin Zhu
- Department of Pharmacology, College of PharmacyHarbin Medical UniversityHarbinHeilongjiangChina
| | - Rui Xie
- Department of Digestive Internal MedicineHarbin Medical University Cancer HospitalHarbinHeilongjiangChina
| |
Collapse
|
72
|
Zheng B, Ding G, Lu G, Li L. Development and external validation of a prognostic nomogram to predict survival in patients aged ≥60 years with pancreatic ductal adenocarcinoma. Transl Cancer Res 2024; 13:2751-2766. [PMID: 38988930 PMCID: PMC11231776 DOI: 10.21037/tcr-24-5] [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: 01/02/2024] [Accepted: 05/07/2024] [Indexed: 07/12/2024]
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC), which accounts for the vast majority of pancreatic cancer (PC), is a highly aggressive malignancy with a dismal prognosis. Age is shown to be an independent factor affecting survival outcomes in patients with PDAC. Our study aimed to identify prognostic factors and construct a nomogram to predict survival in PDAC patients aged ≥60 years. Methods Data of PDAC patients aged ≥60 years were collected from the Surveillance, Epidemiology, and End Results (SEER) database. Multivariate Cox regression analysis was used to determined prognostic factors of overall survival (OS) and cancer-specific survival (CSS), and two nomograms were constructed and validated by calibration plots, concordance index (C-index) and decision curve analysis (DCA). Additionally, 432 patients from the First Affiliated Hospital of Wenzhou Medical University were included as an external cohort. Kaplan-Meier curves were applied to further verify the clinical validity of the nomograms. Results Ten independent prognostic factors were identified to establish the nomograms. The C-indexes of the training and validation groups based on the OS nomogram were 0.759 and 0.760, higher than those of the tumor-node-metastasis (TNM) staging system (0.638 and 0.636, respectively). Calibration curves showed high consistency between predictions and observations. Better area under the receiver operator characteristic (ROC) curve (AUC) values and DCA were also obtained compared to the TNM system. The risk stratification based on the nomogram could distinguish patients with different survival risks. Conclusions We constructed and externally validated a population-based survival-predicting nomogram for PDAC patients aged ≥60 years. The new model could help clinicians personalize survival prediction and risk assessment.
Collapse
Affiliation(s)
- Binjiao Zheng
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Gangfeng Ding
- The First Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Guangrong Lu
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lili Li
- Department of Medical Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
73
|
Marinova M, Khouri DA, Küppers J, Ramig O, Strunk HM, Breuers J, Fazaal J, Fuhrmann C, Coenen M, Möhring C, Zhou T, Zhou X, Anhalt T, Sadeghlar F, Thudium M, Conrad R, Feldmann G, Brossart P, Glowka TR, Kalff JC, Essler M, Strassburg CP, Ko YD, Schmidt-Wolf IGH, Gonzalez-Carmona M. Study Protocol of a Randomized, Two-Arm, Phase I/II Trial Investigating the Feasibility, Safety, and Efficacy of Local Treatment with US-Guided High-Intensity Focused Ultrasound in Combination with Palliative Chemotherapy in Inoperable Pancreatic Cancer. J Clin Med 2024; 13:3717. [PMID: 38999283 PMCID: PMC11242276 DOI: 10.3390/jcm13133717] [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: 05/25/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Pancreatic adenocarcinoma (PaC) still has a dismal prognosis, and despite medical advances, a bleak 5-year survival rate of only 8%, largely due to late diagnosis and limited curative surgical options for most patients. Frontline palliative treatment shows some survival advantages. However, the high disease mortality is accompanied by high morbidity including cancer-related pain and additional symptoms, which strongly impair patients' quality of life (QOL). At present, there is no established strategy for local therapy for PaC primarily aiming to manage local tumor growth and alleviate associated symptoms, particularly pain. In recent years, non-invasive high-intensity focused ultrasound (HIFU) has shown promising results in reducing cancer pain and tumor mass, improving patients' QOL with few side effects. STUDY DESIGN This is the first randomized controlled trial worldwide including 40 patients with inoperable pancreatic adenocarcinoma randomized into two groups: group A undergoing standard chemotherapy; and group B undergoing standard chemotherapy plus local HIFU treatment. This study aims to establish a robust evidence base by examining the feasibility, safety, and efficacy of US-guided HIFU in combination with standard palliative systemic therapy for unresectable PaC. Primary endpoint assessments will focus on parameters including safety issues (phase I), and local response rates (phase II).
Collapse
Affiliation(s)
- Milka Marinova
- Department of Nuclear Medicine, University Hospital Bonn, 53127 Bonn, Germany
| | - David-Alexis Khouri
- Department of Nuclear Medicine, University Hospital Bonn, 53127 Bonn, Germany
| | - Jim Küppers
- Department of Nuclear Medicine, University Hospital Bonn, 53127 Bonn, Germany
| | - Olga Ramig
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, 53127 Bonn, Germany
| | | | - Johannes Breuers
- Department of Nuclear Medicine, University Hospital Bonn, 53127 Bonn, Germany
| | - Julia Fazaal
- Department of Nuclear Medicine, University Hospital Bonn, 53127 Bonn, Germany
| | - Christine Fuhrmann
- D Clinical Study Core Unit Bonn, Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
| | - Martin Coenen
- D Clinical Study Core Unit Bonn, Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
| | - Christian Möhring
- Department of Internal Medicine I, University Hospital Bonn, 53127 Bonn, Germany
| | - Taotao Zhou
- Department of Internal Medicine I, University Hospital Bonn, 53127 Bonn, Germany
| | - Xin Zhou
- Department of Internal Medicine I, University Hospital Bonn, 53127 Bonn, Germany
| | - Thomas Anhalt
- Department of Internal Medicine I, University Hospital Bonn, 53127 Bonn, Germany
| | - Farsaneh Sadeghlar
- Department of Internal Medicine I, University Hospital Bonn, 53127 Bonn, Germany
| | - Marcus Thudium
- Department of Anesthesiology, University Hospital Bonn, 53127 Bonn, Germany
| | - Rupert Conrad
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Muenster, 48149 Muenster, Germany
| | - Georg Feldmann
- Department of Internal Medicine III, University Hospital Bonn, 53127 Bonn, Germany
| | - Peter Brossart
- Department of Internal Medicine III, University Hospital Bonn, 53127 Bonn, Germany
| | - Tim R. Glowka
- Department of Surgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Jörg C. Kalff
- Department of Surgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Markus Essler
- Department of Nuclear Medicine, University Hospital Bonn, 53127 Bonn, Germany
| | | | - Yon-Dschun Ko
- Oncological Center, Johanniter Hospital, 53113 Bonn, Germany
| | | | | |
Collapse
|
74
|
Lu Y, Zhou T, Lu M. A prognostic binary classifier comprised of five critical mRNAs stratified pancreatic cancer patients following resection. Heliyon 2024; 10:e31302. [PMID: 38828350 PMCID: PMC11140619 DOI: 10.1016/j.heliyon.2024.e31302] [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: 11/14/2023] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 06/05/2024] Open
Abstract
Background Pancreatic cancer is characterized by an extremely poor prognosis, even following potentially curative resection. Classical prognostic markers such as histopathological or clinical parameters have limited predictive power. The present study aimed to establish a prognostic model combining mRNA expression data with histopathological and clinical data to better predict survival and stratify pancreatic cancer patients following resection. We pioneered three models in one study and systematically evaluated the clinical benefits of all three models. Methods To identify differentially expressed genes in pancreatic cancer, mRNA data from normal (GTEx database) and pancreatic cancer (TCGA database) tissues were used. Survival analysis was carried out to identify prognosis-relevant genes from the identified differentially expressed genes and LASSO regression was used to filter out hub genes. The risk score of several hub genes was calculated according to gene expression and coefficients. Validation was carried out using an independent set of GEO microarray data. Multivariate COX regression was used for identifying independent clinical and pathological risk factors related to patient's survival in the TCGA database and a prognostic model combining mRNA expression data with histopathological and clinical data was established. Another prognostic model using clinicopathological factors from the SEER database was conceived based on multivariate COX regression. NRI (net reclassification improvement) and IDI (integrated discrimination index) were used to compare the predictive capabilities of the different models. Results We identified 1589 differentially expressed genes (DEGs) through the comparison of normal and pancreatic cancer tissues, of whom 317 were associated with prognosis(p < 0.05). LASSO regression identified five hub genes, MYEOV, ANXA2P2, MET, CEP55, and KRT7, that were used for the five-mRNA-classifier prognostic model. The classifier could stratify patients into a short and long survival group: 5-year overall survival in the training set (TCGA, 6 % vs 52 %, p < 0.001), test set (TCGA, 18 % vs 55 %,p < 0.01) and external validation set (GEO, 0 % vs 25 %, p < 0.05). Sensitivity analysis showed that the mRNA model (model 1) was better than the clinicopathological no-mRNA model (model 2) in predicting 5-year survival in the TCGA database (AUC: 0.877 vs 0.718, z = 3.165, p < 0.01) and better than the multi-factor prognostic model (model 3) from the SEER database (AUC: 0.754, z = 2.637, p < 0.01). On predictive performance, model 1 improved model 2 (NRI = 0.084, z = 1.288, p = 0.198; IDI = 0.055, z = 1.041,p = 0.298) and model 3 (NRI = 0.167,z = 1.961,p = 0.05; IDI = 0.086, z = 1.427, p = 0.154). Conclusion The five-mRNA-classifier is a reliable and feasible instrument to predict the prognosis of pancreatic cancer patients following resection. It might help in patiens counseling and assist clinicians in providing individualized treatment for patients in different risk groups.
Collapse
Affiliation(s)
- Yueqing Lu
- Hepatobiliary and Vascular Surgery, People's Hospital Affiliated to Shandong First Medical University, 271199, Shandong Province, China
| | - Tong Zhou
- Hepatobiliary and Vascular Surgery, People's Hospital Affiliated to Shandong First Medical University, 271199, Shandong Province, China
| | - Mingshu Lu
- Hepatobiliary and Vascular Surgery, People's Hospital Affiliated to Shandong First Medical University, 271199, Shandong Province, China
| |
Collapse
|
75
|
Su J, Wang DS, Hu GX, Liu YY, Hu M, Chen Y, Wang QQ, Yan RC, Wu Y, Li YJ, Ma K, Qi YY, Ding LF, Wu XD. Anti-pancreatic cancer activity of cassane diterpenoids isolated from the seeds of Caesalpinia sappan mediated by autophagy activation via ROS/AMPK/mTORC1 pathway. PHYTOCHEMISTRY 2024; 222:114105. [PMID: 38657886 DOI: 10.1016/j.phytochem.2024.114105] [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: 02/03/2024] [Revised: 04/17/2024] [Accepted: 04/21/2024] [Indexed: 04/26/2024]
Abstract
Three undescribed cassane diterpenoids, caesalpanins D-F (1-3), and seven known ones were isolated from the seeds of Caesalpinia sappan. Structures and absolute configurations of 1-3 were elucidated based on the extensive spectroscopic analysis, single-crystal X-ray diffraction analysis, and ECD calculations. Structurally, compound 1 was the first example of 18-norcassane diterpenoid and 2 was a rare 20-norcassane diterpenoid having an unusual five-membered oxygen bridge between C-10/C-18. The anti-proliferative activity of 1, 3, and 4-10 against PANC-1 cells (pancreatic ductal adenocarcinoma cell line) was evaluated, and phanginin H (4) was found to exhibit anti-cancer activity with IC50 value of 18.13 ± 0.63 μM. Compound 4 inhibited PANC-1 cell growth by arresting the cell cycle at G2/M phase via regulation of cyclin-dependent kinases, and the self-renewal and metastasis of PANC-1 cells by suppressing cancer cell stemness. Furthermore, compound 4 induced ROS generation and subsequently activated autophagy, which was demonstrated by the formation of autophagic vacuoles and dynamic change of autophagic flux. The induced ROS accumulation resulted in AMPK activation and subsequently regulation of mTORC1 activity and ULK phosphorylation, indicating that 4 triggered autophagy through ROS/AMPK/mTORC1 pathway. These findings suggested that 4 might potentially be an autophagy inducer for the therapy of pancreatic cancer.
Collapse
Affiliation(s)
- Jia Su
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, 650500, PR China
| | - De-Sheng Wang
- Key Laboratory of Ethnic Medicine Resource Chemistry, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming, 650504, PR China; School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, PR China
| | - Guo-Xian Hu
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, 650500, PR China
| | - Yu-Yao Liu
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, 650500, PR China
| | - Ming Hu
- Key Laboratory of Ethnic Medicine Resource Chemistry, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming, 650504, PR China
| | - Ya Chen
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, 650500, PR China
| | - Qi-Qi Wang
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, 650500, PR China
| | - Run-Cheng Yan
- Key Laboratory of Ethnic Medicine Resource Chemistry, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming, 650504, PR China
| | - Yun Wu
- Key Laboratory of Ethnic Medicine Resource Chemistry, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming, 650504, PR China
| | - Yong-Jiao Li
- Key Laboratory of Ethnic Medicine Resource Chemistry, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming, 650504, PR China
| | - Ke Ma
- Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, 650011, PR China
| | - Yan-Yan Qi
- Key Laboratory of Ethnic Medicine Resource Chemistry, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming, 650504, PR China.
| | - Lin-Fen Ding
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, PR China; College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, PR China.
| | - Xing-De Wu
- Key Laboratory of Ethnic Medicine Resource Chemistry, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming, 650504, PR China.
| |
Collapse
|
76
|
Schulz B, Leitner E, Schreiber T, Lindner T, Schwarz R, Aboutara N, Ma Y, Escobar HM, Palme R, Hinz B, Vollmar B, Zechner D. Sex Matters-Insights from Testing Drug Efficacy in an Animal Model of Pancreatic Cancer. Cancers (Basel) 2024; 16:1901. [PMID: 38791980 PMCID: PMC11120498 DOI: 10.3390/cancers16101901] [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: 04/27/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Preclinical studies rarely test the efficacy of therapies in both sexes. The field of oncology is no exception in this regard. In a model of syngeneic, orthotopic, metastasized pancreatic ductal adenocarcinoma we evaluated the impact of sex on pathological features of this disease as well as on the efficacy and possible adverse side effects of a novel, small molecule-based therapy inhibiting KRAS:SOS1, MEK1/2 and PI3K signaling in male and female C57BL/6J mice. Male mice had less tumor infiltration of CD8-positive cells, developed bigger tumors, had more lung metastasis and a lower probability of survival compared to female mice. These more severe pathological features in male animals were accompanied by higher distress at the end of the experiment. The evaluated inhibitors BI-3406, trametinib and BKM120 showed synergistic effects in vitro. This combinatorial therapy reduced tumor weight more efficiently in male animals, although the drug concentrations were similar in the tumors of both sexes. These results underline the importance of sex-specific preclinical research and at the same time provide a solid basis for future studies with the tested compounds.
Collapse
Affiliation(s)
- Benjamin Schulz
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (B.S.)
| | - Emily Leitner
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (B.S.)
| | - Tim Schreiber
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (B.S.)
| | - Tobias Lindner
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Center, 18057 Rostock, Germany;
| | - Rico Schwarz
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Nadine Aboutara
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Yixuan Ma
- Department of Medicine Clinic III, Hematology, Oncology and Palliative Medicine, Rostock University Medical Center, 18057 Rostock, Germany
| | - Hugo Murua Escobar
- Department of Medicine Clinic III, Hematology, Oncology and Palliative Medicine, Rostock University Medical Center, 18057 Rostock, Germany
| | - Rupert Palme
- Experimental Endocrinology, Department of Biological Sciences, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Burkhard Hinz
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Brigitte Vollmar
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (B.S.)
| | - Dietmar Zechner
- Rudolf-Zenker-Institute of Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (B.S.)
| |
Collapse
|
77
|
Ren C, Wang Y, Yang X, Tuo Y, Li Y, Gong J. Kikuchi disease: A case report about Sintilimab-induced Kikuchi histiocytic necrotizing lymphadenitis and literature review. Heliyon 2024; 10:e30608. [PMID: 38742085 PMCID: PMC11089371 DOI: 10.1016/j.heliyon.2024.e30608] [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: 08/31/2023] [Revised: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 05/16/2024] Open
Abstract
Immune checkpoint inhibitors have become one of the effective means of solid tumor treatment, among which anti-programmed death-1 (PD-1) antibodies are more maturely applied and can effectively inhibit tumor immune escape, thus enhancing the anti-tumor effect, but it can also lead to a series of immune-related adverse events (irAEs) in the process of clinical use. Here, we report a Patient with pancreatic solid pseudopapilloma treated with Sintilimab for the fifteenth cycles who developed chills, fever, and lymph node enlargement. Considering that the patient did not have infection, without history of autoimmune disease, we diagnosed the patient with Sintilimab-induced histiocytic necrotizing lymphadenitis (Kikuchi disease). The symptoms are alleviated after rapid use of glucocorticoids. Histiocytic necrotizing lymphadenitis (Kikuchi lymphadenitis) with anti-programmed death-1 (PD-1) antibody is a rare immune-related adverse events (irAEs).
Collapse
Affiliation(s)
- Chunxiao Ren
- Department of Hematology, Dazhou Central Hospital, Tongchuan District, 56 Nanyue Temple Street Sichuan, Dazhou, 635000, China
| | - Yuqun Wang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, 261042, China
| | - Xin Yang
- Department of Hematology, Dazhou Central Hospital, Tongchuan District, 56 Nanyue Temple Street Sichuan, Dazhou, 635000, China
| | - Yinglan Tuo
- Pathology Department, Dazhou Central Hospital, Tongchuan District, 56 Nanyue Temple Street Sichuan, Dazhou, 635000, China
| | - Yaqiong Li
- Department of Hematology, Dazhou Central Hospital, Tongchuan District, 56 Nanyue Temple Street Sichuan, Dazhou, 635000, China
| | - Jichang Gong
- Department of Hematology, Dazhou Central Hospital, Tongchuan District, 56 Nanyue Temple Street Sichuan, Dazhou, 635000, China
| |
Collapse
|
78
|
Long SA, Amparo AM, Goodhart G, Ahmad SA, Waters AM. Evaluation of KRAS inhibitor-directed therapies for pancreatic cancer treatment. Front Oncol 2024; 14:1402128. [PMID: 38800401 PMCID: PMC11116577 DOI: 10.3389/fonc.2024.1402128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
Despite significant advancements in the treatment of other cancers, pancreatic ductal adenocarcinoma (PDAC) remains one of the world's deadliest cancers. More than 90% of PDAC patients harbor a Kirsten rat sarcoma (KRAS) gene mutation. Although the clinical potential of anti-KRAS therapies has long been realized, all initial efforts to target KRAS were unsuccessful. However, with the recent development of a new generation of KRAS-targeting drugs, multiple KRAS-targeted treatment options for patients with PDAC have entered clinical trials. In this review, we provide an overview of current standard of care treatment, describe RAS signaling and the relevance of KRAS mutations, and discuss RAS isoform- and mutation-specific differences. We also evaluate the clinical efficacy and safety of mutation-selective and multi-selective inhibitors, in the context of PDAC. We then provide a comparison of clinically relevant KRAS inhibitors to second-line PDAC treatment options. Finally, we discuss putative resistance mechanisms that may limit the clinical effectiveness of KRAS-targeted therapies and provide a brief overview of promising therapeutic approaches in development that are focused on mitigating these resistance mechanisms.
Collapse
Affiliation(s)
- Szu-Aun Long
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Amber M. Amparo
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Grace Goodhart
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Syed A. Ahmad
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Andrew M. Waters
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
79
|
Zhuang J, Wang S, Wang Y, Wu Y, Hu R. Prognostic significance of preoperative lymphocytes, albumin, and neutrophils (LANR) index in resectable pancreatic ductal adenocarcinoma. BMC Cancer 2024; 24:568. [PMID: 38714979 PMCID: PMC11075219 DOI: 10.1186/s12885-024-12329-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/02/2024] [Indexed: 05/12/2024] Open
Abstract
PURPOSE The index composed of preoperative lymphocytes, albumin, and neutrophils (LANR), a new composite score based on inflammatory response and nutritional status, has been reported to be associated with the prognosis of multiple types of cancer, but the role of LANR in the prognosis of resectable pancreatic ductal adenocarcinoma (PDAC) has not yet been elucidated. PATIENTS AND METHODS The data of 142 patients with PDAC who underwent radical resection in the Affiliated Hospital of Jiangnan University from January 2015 to December 2018 were retrospectively analyzed. Receiver Operating Characteristic (ROC) curves were generated to determine the optimal cut-off values for these parameters, as well as the sensitivity and specificity of LANR in predicting survival. The Kaplan-Meier method was used to draw the survival curves. Log rank test was used for univariate analysis, and Cox proportional hazards regression model was used for multivariate analysis. RESULTS: The optimal cut-off value of LANR was 18.145, and a low preoperative LANR was significantly correlated with the location of the tumor (p = 0.047). Multivariate analysis showed that tumor differentiation degree (HR:2.357, 95%CI:1.388-4.003,p = 0.002), lymph node metastasis (HR:1.755, 95%CI: 1.115-2.763, p = 0.015), TNM stage (HR:4.686, 95%CI: 2.958-7.425, p < 0.001), preoperative cancer antigen 19 - 9 levels (HR:1.001, 95%CI: 1.000-1.001, p < 0.001) and preoperative LANR (HR:0.221, 95%CI: 0.111-0.441, p < 0.001) were independent risk factors for a poor prognosis in patients undergoing radical resection of PDAC. CONCLUSION This study found that preoperative LANR can be used to assess the prognosis of radical resection in patients with PDAC; those with low preoperative LANR had a worse outcome.
Collapse
Affiliation(s)
- Jiaru Zhuang
- Department of Laboratory Medicine, Jiangnan University Medical Center (Wuxi No People's Hospital), 68 Zhongshan Road, Wuxi, Jiangsu, 214000, China
| | - Shan Wang
- Human reproductive medicine center, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, Jiangsu, 214026, China
| | - Yuan Wang
- Human reproductive medicine center, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, Jiangsu, 214026, China
| | - Yibo Wu
- Human reproductive medicine center, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, Jiangsu, 214026, China.
| | - Renjing Hu
- Department of Laboratory Medicine, Jiangnan University Medical Center (Wuxi No People's Hospital), 68 Zhongshan Road, Wuxi, Jiangsu, 214000, China.
| |
Collapse
|
80
|
Chien MS, Lin CC, Lai JH. Endoscopic Ultrasonography-Guided Fine-Needle Biopsy for Patients with Resectable Pancreatic Malignancies. GASTROENTEROLOGY INSIGHTS 2024; 15:375-385. [DOI: 10.3390/gastroent15020026] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Clinicians often use endoscopic ultrasonography to survey pancreatic tumors. When endoscopists conduct this examination and find the tumor to be unresectable, a fine-needle biopsy is subsequently performed for tissue confirmation. However, if the tumor is deemed resectable, the necessity of a pre-operative fine-needle biopsy remains debatable. Therefore, we performed a retrospective analysis of a single-center cohort of patients with pancreatic tumors who underwent an endoscopic ultrasound-guided fine-needle biopsy or aspiration (EUS-FNB or FNA) between 2020 and 2022. This study focused on patients diagnosed with resectable malignant pancreatic tumors. The exclusion criteria included individuals diagnosed with benign pancreatic lesions and those with unresectable tumors. A total of 68 patients were enrolled in this study. Histological examination revealed that pancreatic adenocarcinoma was the predominant type of tumor (n = 42, 61.8%), followed by neuroendocrine tumors (n = 22, 32.3%), and metastasis (n = 4, 5.9%). Notably, 17 patients had a history of other cancers, with 23.5% being diagnosed with a metastatic tumor rather than primary pancreatic cancer. Therefore, EUS-FNA/FNB is crucial in patients with a resectable pancreatic tumor and a history of cancer to differentiate between a primary and a metastatic tumor.
Collapse
Affiliation(s)
- Ming-Sheng Chien
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 104217, Taiwan
| | - Ching-Chung Lin
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 104217, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
| | - Jian-Han Lai
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 104217, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Medicine Nursing and Management, Taipei 11260, Taiwan
| |
Collapse
|
81
|
Li F, Si W, Xia L, Yin D, Wei T, Tao M, Cui X, Yang J, Hong T, Wei R. Positive feedback regulation between glycolysis and histone lactylation drives oncogenesis in pancreatic ductal adenocarcinoma. Mol Cancer 2024; 23:90. [PMID: 38711083 PMCID: PMC11071201 DOI: 10.1186/s12943-024-02008-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/24/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Metabolic reprogramming and epigenetic alterations contribute to the aggressiveness of pancreatic ductal adenocarcinoma (PDAC). Lactate-dependent histone modification is a new type of histone mark, which links glycolysis metabolite to the epigenetic process of lactylation. However, the role of histone lactylation in PDAC remains unclear. METHODS The level of histone lactylation in PDAC was identified by western blot and immunohistochemistry, and its relationship with the overall survival was evaluated using a Kaplan-Meier survival plot. The participation of histone lactylation in the growth and progression of PDAC was confirmed through inhibition of histone lactylation by glycolysis inhibitors or lactate dehydrogenase A (LDHA) knockdown both in vitro and in vivo. The potential writers and erasers of histone lactylation in PDAC were identified by western blot and functional experiments. The potential target genes of H3K18 lactylation (H3K18la) were screened by CUT&Tag and RNA-seq analyses. The candidate target genes TTK protein kinase (TTK) and BUB1 mitotic checkpoint serine/threonine kinase B (BUB1B) were validated through ChIP-qPCR, RT-qPCR and western blot analyses. Next, the effects of these two genes in PDAC were confirmed by knockdown or overexpression. The interaction between TTK and LDHA was identified by Co-IP assay. RESULTS Histone lactylation, especially H3K18la level was elevated in PDAC, and the high level of H3K18la was associated with poor prognosis. The suppression of glycolytic activity by different kinds of inhibitors or LDHA knockdown contributed to the anti-tumor effects of PDAC in vitro and in vivo. E1A binding protein p300 (P300) and histone deacetylase 2 were the potential writer and eraser of histone lactylation in PDAC cells, respectively. H3K18la was enriched at the promoters and activated the transcription of mitotic checkpoint regulators TTK and BUB1B. Interestingly, TTK and BUB1B could elevate the expression of P300 which in turn increased glycolysis. Moreover, TTK phosphorylated LDHA at tyrosine 239 (Y239) and activated LDHA, and subsequently upregulated lactate and H3K18la levels. CONCLUSIONS The glycolysis-H3K18la-TTK/BUB1B positive feedback loop exacerbates dysfunction in PDAC. These findings delivered a new exploration and significant inter-relationship between lactate metabolic reprogramming and epigenetic regulation, which might pave the way toward novel lactylation treatment strategies in PDAC therapy.
Collapse
Affiliation(s)
- Fei Li
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China
| | - Wenzhe Si
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Li Xia
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China
| | - Deshan Yin
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China
| | - Tianjiao Wei
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China
| | - Ming Tao
- Department of General Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Xiaona Cui
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China
| | - Jin Yang
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China
| | - Tianpei Hong
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China.
| | - Rui Wei
- Department of Endocrinology and Metabolism, State Key Laboratory of Female Fertility Promotion, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
82
|
Zhang J, Teng F, Hu B, Liu W, Huang Y, Wu J, Wang Y, Su H, Yang S, Zhang L, Guo L, Lei Z, Yan M, Xu X, Wang R, Bao Q, Dong Q, Long J, Qian K. Early Diagnosis and Prognosis Prediction of Pancreatic Cancer Using Engineered Hybrid Core-Shells in Laser Desorption/Ionization Mass Spectrometry. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311431. [PMID: 38241281 DOI: 10.1002/adma.202311431] [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: 10/31/2023] [Revised: 01/11/2024] [Indexed: 01/21/2024]
Abstract
Effective detection of bio-molecules relies on the precise design and preparation of materials, particularly in laser desorption/ionization mass spectrometry (LDI-MS). Despite significant advancements in substrate materials, the performance of single-structured substrates remains suboptimal for LDI-MS analysis of complex systems. Herein, designer Au@SiO2@ZrO2 core-shell substrates are developed for LDI-MS-based early diagnosis and prognosis of pancreatic cancer (PC). Through controlling Au core size and ZrO2 shell crystallization, signal amplification of metabolites up to 3 orders is not only achieved, but also the synergistic mechanism of the LDI process is revealed. The optimized Au@SiO2@ZrO2 enables a direct record of serum metabolic fingerprints (SMFs) by LDI-MS. Subsequently, SMFs are employed to distinguish early PC (stage I/II) from controls, with an accuracy of 92%. Moreover, a prognostic prediction scoring system is established with enhanced efficacy in predicting PC survival compared to CA19-9 (p < 0.05). This work contributes to material-based cancer diagnosis and prognosis.
Collapse
Affiliation(s)
- Juxiang Zhang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Fei Teng
- Department of Gastrointestinal Surgery, Minhang Hospital, Fudan University, Shanghai, 201199, China
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Beiyuan Hu
- Department of Pancreatic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Wanshan Liu
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yida Huang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jiao Wu
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yuning Wang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Haiyang Su
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Shouzhi Yang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Lumin Zhang
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Lingchuan Guo
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Zhe Lei
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Meng Yan
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Xiaoyu Xu
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Ruimin Wang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Qingui Bao
- Fosun Diagnostics (Shanghai) Co., Ltd, Shanghai, 200435, China
| | - Qiongzhu Dong
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Jiang Long
- Department of Pancreatic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Kun Qian
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| |
Collapse
|
83
|
Shimizu T, Maeda S, Link J, Deranteriassian A, Premji A, Verma A, Chervu N, Park J, Girgis M, Benharash P, Hines J, Wainberg Z, Wolfgang C, Burns W, Yu J, Fernandez-Del Castillo C, Lillemoe K, Ferrone C, Donahue T. Clinical and pathological factors associated with survival in patients with pancreatic cancer who receive adjuvant therapy after neoadjuvant therapy: A retrospective multi-institutional analysis. Surgery 2024; 175:1377-1385. [PMID: 38365548 DOI: 10.1016/j.surg.2024.01.008] [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: 06/02/2023] [Revised: 12/29/2023] [Accepted: 01/08/2024] [Indexed: 02/18/2024]
Abstract
BACKGROUND Neoadjuvant therapy is being increasingly used for patients with pancreatic cancer. The role of adjuvant therapy in these patients is unclear. The purpose of this study was to identify clinical and pathologic characteristics that are associated with longer overall survival in patients with pancreatic cancer who receive adjuvant therapy after neoadjuvant therapy. METHODS This study was conducted using multi-institutional data. All patients underwent surgery after at least 1 cycle of neoadjuvant therapy for pancreatic cancer. Patients who died within 3 months after surgery and were known to have distant metastasis or macroscopic residual disease were excluded. Mann-Whitney U test, χ2 analysis, Kaplan-Meier plot, and univariate and multivariate Cox regression analysis were performed as statistical analyses. RESULTS In the present study, 529 patients with resected pancreatic cancer after neoadjuvant therapy were reviewed. For neoadjuvant therapy, 177 (33.5%) patients received neoadjuvant chemotherapy, and 352 (66.5%) patients received neoadjuvant chemoradiotherapy. The median duration of neoadjuvant therapy was 7.0 months (interquartile range, 5.0-8.7). Patients were followed for a median of 23.0 months after surgery. Adjuvant therapy was administered to 297 (56.1%) patients and was not associated with longer overall survival for the entire cohort (24 vs 22 months, P = .31). Interaction analysis showed that adjuvant therapy was associated with longer overall survival in patients who received less than 4 months neoadjuvant therapy (hazard ratio 0.40; 95% confidence interval 0.17-0.95; P = .03) or who had microscopic margin positive surgical resections (hazard ratio 0.56; 95% confidence interval 0.33-0.93; P = .03). CONCLUSION In this retrospective study, there was a survival benefit associated with adjuvant therapy for patients who received less than 4 months of neoadjuvant therapy or had microscopic positive margins.
Collapse
Affiliation(s)
- Takayuki Shimizu
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Shimpei Maeda
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Jason Link
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | | | - Alykhan Premji
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Arjun Verma
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Nikhil Chervu
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Joon Park
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Mark Girgis
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Peyman Benharash
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Joe Hines
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Zev Wainberg
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Christopher Wolfgang
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - William Burns
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jun Yu
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Keith Lillemoe
- Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - Cristina Ferrone
- Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - Timothy Donahue
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA.
| |
Collapse
|
84
|
Damare R, Engle K, Kumar G. Targeting epidermal growth factor receptor and its downstream signaling pathways by natural products: A mechanistic insight. Phytother Res 2024; 38:2406-2447. [PMID: 38433568 DOI: 10.1002/ptr.8166] [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: 08/02/2023] [Revised: 01/30/2024] [Accepted: 02/03/2024] [Indexed: 03/05/2024]
Abstract
The epidermal growth factor receptor (EGFR) is a transmembrane receptor tyrosine kinase (RTK) that maintains normal tissues and cell signaling pathways. EGFR is overactivated and overexpressed in many malignancies, including breast, lung, pancreatic, and kidney. Further, the EGFR gene mutations and protein overexpression activate downstream signaling pathways in cancerous cells, stimulating the growth, survival, resistance to apoptosis, and progression of tumors. Anti-EGFR therapy is the potential approach for treating malignancies and has demonstrated clinical success in treating specific cancers. The recent report suggests most of the clinically used EGFR tyrosine kinase inhibitors developed resistance to the cancer cells. This perspective provides a brief overview of EGFR and its implications in cancer. We have summarized natural products-derived anticancer compounds with the mechanistic basis of tumor inhibition via the EGFR pathway. We propose that developing natural lead molecules into new anticancer agents has a bright future after clinical investigation.
Collapse
Affiliation(s)
- Rutuja Damare
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, India
| | - Kritika Engle
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, India
| | - Gautam Kumar
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, India
| |
Collapse
|
85
|
Frimpong E, Bulusu R, Okoro J, Inkoom A, Ndemazie N, Rogers S, Zhu X, Han B, Agyare E. Development of novel pyrimidine nucleoside analogs as potential anticancer agents: Synthesis, characterization, and In-vitro evaluation against pancreatic cancer. Eur J Pharm Sci 2024; 196:106754. [PMID: 38554983 PMCID: PMC11229414 DOI: 10.1016/j.ejps.2024.106754] [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: 01/24/2024] [Revised: 03/18/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024]
Abstract
The present study proposed modification of 5-FU by conjugation with an acyl chloride and a 5-membered heterocyclic ring to improve its in-vitro cytotoxicity and metabolic stability. XYZ-I-71 and XYZ-I-73 were synthesized by introducing a tetrahydrofuran ring on 5-fluorocytosine (a precursor of 5-FU) and conjugation with octanoyl chloride and lauroyl chloride, respectively. The structure of the synthesized compounds was validated using NMR and micro-elemental analysis. The antiproliferative activity of the analogs was determined against MiaPaCa-2, PANC-1, and BxPC-3 pancreatic cancer cells. The analog's stability in human liver microsomes was quantified by HPLC. We found that the XYZ-I-73 (IC50 3.6 ± 0.4 μM) analog was most effective against MiaPaCa-2 cells compared to XYZ-I-71(IC50 12.3 ± 1.7 μM), GemHCl (IC50 24.2 ± 1.3 μM), Irinotecan (IC50 10.1 ± 1.5 μM) and 5-FU (IC50 13.2 ± 1.1 μM). The antiproliferative effects of this analog in Miapaca-2 cells is evident based on it having a 7-fold,3-fold, and 4-fold increased cytotoxic effect over Gem-HCl, Irinotecan, and 5-FU, respectively. On the other hand, XYZ-I-71 exhibited a 2-fold increased cytotoxic effect over Gem-HCl but a comparable cytotoxic effect to 5-FU and Irinotecan in MiaPaCa-2 cells. A similar trend of higher XYZ-I-73 inhibition was observed in PANC-1 and BxPC-3 cultures. For 48-h MiaPaCa-2 cell migration studies, XYZ-I-73 (5 μM) significantly reduced migration (# of migrated cells, 168 ± 2.9), followed by XYZ-I-71(315±2.1), Gem-HCl (762±3.1) and 5-FU (710 ± 3.2). PARP absorbance studies demonstrated significant inhibition of PARP expression of XYZ-I-73 treated cells compared to 5-FU, GemHCl, and XYZ-I-71. Further, BAX and p53 expressions were significantly increased in cells treated with XYZ-I-73 compared to 5-FU, GemHCl, and XYZ-I-71. In-vitro, metabolic stability studies showed that 80 ± 5.9% of XYZ-I-71 and XYZ-I-73 remained intact after 2 h exposure in liver microsomal solution compared to 5-FU. The XYZ-I-73 analog demonstrated a remarkable cytotoxic effect and improved in-vitro metabolic stability over the selected standard drugs and may have potential anticancer activity against pancreatic cancer.
Collapse
Affiliation(s)
- Esther Frimpong
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Raviteja Bulusu
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Joy Okoro
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Andriana Inkoom
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Nkafu Ndemazie
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States; Department of Internal Medicine, Richmond University Medical Center, Staten Island, NY, United States
| | - Sherise Rogers
- Department of Medicine, Division of Hematology and Oncology, University of Florida College of Medicine, Gainesville, FL, United States
| | - Xue Zhu
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Bo Han
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Edward Agyare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States.
| |
Collapse
|
86
|
Kang MJ, Li HX, Gan Y, Fang C, Yang XL, Li B, Su S. Efficacy and safety of first-line chemotherapies for patients with advanced pancreatic ductal adenocarcinoma: A systematic review and network meta-analysis. Heliyon 2024; 10:e27679. [PMID: 38681566 PMCID: PMC11046077 DOI: 10.1016/j.heliyon.2024.e27679] [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: 04/08/2023] [Revised: 02/05/2024] [Accepted: 03/05/2024] [Indexed: 05/01/2024] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease, often diagnosed at an advanced stage. Systemic chemotherapy is the primary treatment, but direct comparisons of different regimens are limited. This study conducted a systematic review and network meta-analysis (NMA) to compare the efficacy and safety of various chemotherapy regimens, with the unique advantage of only including Phase III randomized controlled trials (RCTs). Methods NMA was conducted regarding the searched phase III RCTs by comparing overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and adverse events (AEs) of different chemotherapy protocols. Results The analysis included 24 studies with 11470 patients across 25 treatment modalities. Among the chemotherapy regimens evaluated, FOLFIRINOX (fluorouracil, leucovorin, irinotecan, and oxaliplatin) demonstrated the highest OS and PFS, with a risk ratio (logHR) of 4.5 (95 % confidence interval 4.32-4.68) compared to gemcitabine monotherapy. The PEFG regimen (cisplatin, epirubicin, 5-fluorouracil, and gemcitabine) exhibited the highest ORR, with an odds ratio (OR) of 6.67 (2.08-20) compared to gemcitabine monotherapy. Notably, gemcitabine plus sorafenib was associated with the lowest hematological toxicity, with an odds ratio (OR) of 0.1 (0.02-0.48). Conclusion Combination therapies may offer greater benefits but also cause more toxic effects. However, combinations with targeted agents seem to have fewer adverse reactions.
Collapse
Affiliation(s)
| | | | - Yu Gan
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Cheng Fang
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Xiao-Li Yang
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Bo Li
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Song Su
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Sichuan, China
| |
Collapse
|
87
|
Liou GY, Byrd CJ, Storz P, Messex JK. Cytokine CCL9 Mediates Oncogenic KRAS-Induced Pancreatic Acinar-to-Ductal Metaplasia by Promoting Reactive Oxygen Species and Metalloproteinases. Int J Mol Sci 2024; 25:4726. [PMID: 38731942 PMCID: PMC11083758 DOI: 10.3390/ijms25094726] [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: 02/29/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) can originate from acinar-to-ductal metaplasia (ADM). Pancreatic acini harboring oncogenic Kras mutations are transdifferentiated to a duct-like phenotype that further progresses to become pancreatic intraepithelial neoplasia (PanIN) lesions, giving rise to PDAC. Although ADM formation is frequently observed in KrasG12D transgenic mouse models of PDAC, the exact mechanisms of how oncogenic KrasG12D regulates this process remain an enigma. Herein, we revealed a new downstream target of oncogenic Kras, cytokine CCL9, during ADM formation. Higher levels of CCL9 and its receptors, CCR1 and CCR3, were detected in ADM regions of the pancreas in p48cre:KrasG12D mice and human PDAC patients. Knockdown of CCL9 in KrasG12D-expressed pancreatic acini reduced KrasG12D-induced ADM in a 3D organoid culture system. Moreover, exogenously added recombinant CCL9 and overexpression of CCL9 in primary pancreatic acini induced pancreatic ADM. We also showed that, functioning as a downstream target of KrasG12D, CCL9 promoted pancreatic ADM through upregulation of the intracellular levels of reactive oxygen species (ROS) and metalloproteinases (MMPs), including MMP14, MMP3 and MMP2. Blockade of MMPs via its generic inhibitor GM6001 or knockdown of specific MMP such as MMP14 and MMP3 decreased CCL9-induced pancreatic ADM. In p48cre:KrasG12D transgenic mice, blockade of CCL9 through its specific neutralizing antibody attenuated pancreatic ADM structures and PanIN lesion formation. Furthermore, it also diminished infiltrating macrophages and expression of MMP14, MMP3 and MMP2 in the ADM areas. Altogether, our results provide novel mechanistic insight into how oncogenic Kras enhances pancreatic ADM through its new downstream target molecule, CCL9, to initiate PDAC.
Collapse
Affiliation(s)
- Geou-Yarh Liou
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
- Department of Biological Sciences, Clark Atlanta University, Atlanta, GA 30314, USA
| | - Crystal J. Byrd
- Department of Biological Sciences, Clark Atlanta University, Atlanta, GA 30314, USA
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Justin K. Messex
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
| |
Collapse
|
88
|
Holze M, Loos M, Hüttner F, Tenckhoff S, Feisst M, Knebel P, Klotz R, Mehrabi A, Michalski C, Pianka F. Cavitron ultrasonic surgical aspirator (CUSA) compared with conventional pancreatic transection in distal pancreatectomy: study protocol for the randomised controlled CUSA-1 pilot trial. BMJ Open 2024; 14:e082024. [PMID: 38637127 PMCID: PMC11029322 DOI: 10.1136/bmjopen-2023-082024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 03/08/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Postoperative pancreatic fistula (POPF) remains the most common and serious complication after distal pancreatectomy. Many attempts at lowering fistula rates have led to unrewarding insignificant results as still up to 30% of the patients suffer from clinically relevant POPF. Therefore, the development of new innovative methods and procedures is still a cornerstone of current surgical research.The cavitron ultrasonic surgical aspirator (CUSA) device is a well-known ultrasound-based parenchyma transection method, often used in liver and neurosurgery which has not yet been thoroughly investigated in pancreatic surgery, but the first results seem very promising. METHODS The CUSA-1 trial is a randomised controlled pilot trial with two parallel study groups. This single-centre trial is assessor and patient blinded. A total of 60 patients with an indication for open distal pancreatectomy will be intraoperatively randomised after informed consent. The patients will be randomly assigned to either the control group with conventional pancreas transection (scalpel or stapler) or the experimental group, with transection using the CUSA device. The primary safety endpoint of this trial will be postoperative complications ≥grade 3 according to the Clavien-Dindo classification. The primary endpoint to investigate the effect will be the rate of POPF within 30 days postoperatively according to the ISGPS definition. Further perioperative outcomes, including postpancreatectomy haemorrhage, length of hospital stay and mortality will be analysed as secondary endpoints. DISCUSSION Based on the available literature, CUSA may have a beneficial effect on POPF occurrence after distal pancreatectomy. The rationale of the CUSA-1 pilot trial is to investigate the safety and feasibility of the CUSA device in elective open distal pancreatectomy compared with conventional dissection methods and gather the first data on the effect on POPF occurrence. This data will lay the groundwork for a future confirmatory multicentre randomised controlled trial. ETHICS AND DISSEMINATION The CUSA-1 trial protocol was approved by the ethics committee of the University of Heidelberg (No. S-098/2022). Results will be published in an international peer-reviewed journal and summaries will be provided in lay language to study participants and their relatives. TRIAL REGISTRATION NUMBER DRKS00027474.
Collapse
Affiliation(s)
- Magdalena Holze
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
- SDGC, The Study Centre of the German Surgical Society, Heidelberg, Germany
| | - Martin Loos
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Felix Hüttner
- Klinik für Allgemein-, Viszeral- und Thoraxchirurgie, Klinikum Nürnberg, Nurnberg, Germany
| | - Solveig Tenckhoff
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
- SDGC, The Study Centre of the German Surgical Society, Heidelberg, Germany
| | - Manuel Feisst
- Institute for Medical Biometry, University of Heidelberg, Heidelberg, Germany
| | - Phillip Knebel
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Rosa Klotz
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
- SDGC, The Study Centre of the German Surgical Society, Heidelberg, Germany
| | - Arianeb Mehrabi
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Christoph Michalski
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Frank Pianka
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
- SDGC, The Study Centre of the German Surgical Society, Heidelberg, Germany
| |
Collapse
|
89
|
Muaddi H, Kearse L, Warner S. Multimodal Approaches to Patient Selection for Pancreas Cancer Surgery. Curr Oncol 2024; 31:2260-2273. [PMID: 38668070 PMCID: PMC11049254 DOI: 10.3390/curroncol31040167] [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: 03/24/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
With an overall 5-year survival rate of 12%, pancreas ductal adenocarcinoma (PDAC) is an aggressive cancer that claims more than 50,000 patient lives each year in the United States alone. Even those few patients who undergo curative-intent resection with favorable pathology reports are likely to experience recurrence within the first two years after surgery and ultimately die from their cancer. We hypothesize that risk factors for these early recurrences can be identified with thorough preoperative staging, thus enabling proper patient selection for surgical resection and avoiding unnecessary harm. Herein, we review evidence supporting multidisciplinary and multimodality staging, comprehensive neoadjuvant treatment strategies, and optimal patient selection for curative-intent surgical resections. We further review data generated from our standardized approach at the Mayo Clinic and extrapolate to inform potential future investigations.
Collapse
Affiliation(s)
| | | | - Susanne Warner
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MN 55902, USA; (H.M.)
| |
Collapse
|
90
|
Xu M, Tang J, Sun Q, Meng J, Chen G, Chang Y, Yao Y, Ji J, Luo H, Chen L, Lu M, Shi W. CENPN contributes to pancreatic carcinoma progression through the MDM2-mediated p53 signaling pathway. Arch Med Sci 2024; 20:1655-1671. [PMID: 39649279 PMCID: PMC11623148 DOI: 10.5114/aoms/171956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 09/06/2023] [Indexed: 12/10/2024] Open
Abstract
Introduction We undertook an in-depth investigation of the data pertaining to pancreatic adenocarcinoma (PAAD) to identify potential targets for the development of precision therapies. Material and methods The construction of a protein-protein interaction (PPI) network was based on overlapping differentially expressed genes (DEGs) identified in the GSE16515, GSE32676, and GSE125158 datasets. A subsequent bioinformatic analysis was performed on the interconnected genes within the PPI network, leading to the identification of the central gene, CENPN. In vitro experimentation such as CCK8 and Transwell experiments was employed to elucidate the impact of CENPN expression patterns on PAAD cell proliferation, migration, and invasion. Furthermore, the investigation revealed through comprehensive enrichment analysis that the pivotal signaling pathway associated with CENPN is the p53 signaling pathway. Results Following a comprehensive bioinformatic analysis of 161 concordant differentially expressed genes (DEGs) across three microarray datasets, CENPN emerged as the central gene under investigation. Overexpression of CENPN in pancreatic adenocarcinoma (PAAD) was associated with unfavorable patient outcomes and heightened sensitivity to four PAAD therapies: gemcitabine, docetaxel, paclitaxel, and sunitinib. Reduced CENPN expression impeded PAAD cell proliferation, migration, and invasion; however, these effects were counteracted upon upregulation of CENPN expression. Additionally, CENPN interacted with MDM2, promoting PAAD progression by targeting the p53 signaling pathway. Conclusions The findings of our study substantiate that CENPN is associated with the pathogenesis of PAAD. Consequently, CENPN appears to be a promising candidate for targeted precision therapy in clinical applications.
Collapse
Affiliation(s)
- Ming Xu
- Department of Gastroenterology, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Jie Tang
- Department of Gastroenterology, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Qiong Sun
- Department of Oncology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jing Meng
- Department of Oncology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Guoyu Chen
- Department of Gastroenterology, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Yunli Chang
- Department of Gastroenterology, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Yao Yao
- Department of Gastroenterology, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Jieru Ji
- Department of Gastroenterology, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Hao Luo
- Department of Gastroenterology, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Lingling Chen
- Department of Gastroenterology, Shanghai Pudong New Area People’s Hospital, Shanghai, China
| | - Minxue Lu
- Department of Gastroenterology, Huzhou College Affiliated Nantaihu Hospital, Huzhou, Zhejiang, China
| | - Weiwei Shi
- Department of Oncology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
91
|
Li Z, Mo F, Guo K, Ren S, Wang Y, Chen Y, Schwartz PB, Richmond N, Liu F, Ronnekleiv-Kelly SM, Hu Q. Nanodrug-bacteria conjugates-mediated oncogenic collagen depletion enhances immune checkpoint blockade therapy against pancreatic cancer. MED 2024; 5:348-367.e7. [PMID: 38521069 DOI: 10.1016/j.medj.2024.02.012] [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: 06/25/2023] [Revised: 11/15/2023] [Accepted: 02/27/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) cancer cells specifically produce abnormal oncogenic collagen to bind with integrin α3β1 receptor and activate the downstream focal adhesion kinase (FAK), protein kinase B (AKT), and mitogen-activated protein kinase (MAPK) signaling pathway. Collectively, this promotes immunosuppression and tumor proliferation and restricts the response rate of clinical cancer immunotherapies. METHODS Here, by leveraging the hypoxia tropism and excellent motility of the probiotic Escherichia coli strain Nissle 1917 (ECN), we developed nanodrug-bacteria conjugates to penetrate the extracellular matrix (ECM) and shuttle the surface-conjugated protein cages composed of collagenases and anti-programmed death-ligand 1 (PD-L1) antibodies to PDAC tumor parenchyma. FINDINGS We found the oncogenic collagen expression in human pancreatic cancer patients and demonstrated its interaction with integrin α3β1. We proved that reactive oxygen species (ROS) in the microenvironment of PDAC triggered collagenase release to degrade oncogenic collagen and block integrin α3β1-FAK signaling pathway, thus overcoming the immunosuppression and synergizing with anti-PD-L1 immunotherapy. CONCLUSIONS Collectively, our study highlights the significance of oncogenic collagen in PDAC immunotherapy, and consequently, we developed a therapeutic strategy that can deplete oncogenic collagen to synergize with immune checkpoint blockade for enhanced PDAC treatment efficacy. FUNDING This work was supported by the University of Wisconsin Carbone Cancer Center Research Collaborative and Pancreas Cancer Research Task Force, UWCCC Transdisciplinary Cancer Immunology-Immunotherapy Pilot Project, and the start-up package from the University of Wisconsin-Madison (to Q.H.).
Collapse
Affiliation(s)
- Zhaoting Li
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA; Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Fanyi Mo
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kai Guo
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Shuai Ren
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Yixin Wang
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA; Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yu Chen
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA; Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Patrick B Schwartz
- Department of Surgery, Division of Surgical Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Nathaniel Richmond
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Fengyuan Liu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Sean M Ronnekleiv-Kelly
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Surgery, Division of Surgical Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA; Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
92
|
Chen T, Mao S, Ma J, Tang X, Zhu R, Mao D, Zhu X, Pan Q. Proximity-Enhanced Functional Imaging Analysis of Engineered Tumors. Angew Chem Int Ed Engl 2024; 63:e202319117. [PMID: 38305848 DOI: 10.1002/anie.202319117] [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: 12/12/2023] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 02/03/2024]
Abstract
Functional imaging (FI) techniques have revolutionized tumor imaging by providing information on specific tumor functions, such as glycometabolism. However, tumor cells lack unique molecular characteristics at the molecular level and metabolic pathways, resulting in limited metabolic differences compared to normal cells and increased background signals from FI. To address this limitation, we developed a novel imaging technique termed proximity-enhanced functional imaging (PEFI) for accurate visualization of tumors. By using "two adjacent chemically labeled glycoproteins" as output signals, we significantly enhance the metabolic differences between tumor and normal cells by PEFI, thereby reducing the background signals for analysis and improving the accuracy of tumor functional imaging. Our results demonstrate that PEFI can accurately identify tumors at the cellular, tissue, and animal level, and has potential value in clinical identification and analysis of tumor cells and tissues, as well as in the guidance of clinical tumor resection surgery.
Collapse
Affiliation(s)
- Tianshu Chen
- Clinical Laboratory, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Shanghai Key Laboratory of Clinical Molecular Diagnostics for Pediatrics, Shanghai, 200127, P. R. China
| | - Siwei Mao
- Clinical Laboratory, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Shanghai Key Laboratory of Clinical Molecular Diagnostics for Pediatrics, Shanghai, 200127, P. R. China
| | - Ji Ma
- Clinical Laboratory, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Shanghai Key Laboratory of Clinical Molecular Diagnostics for Pediatrics, Shanghai, 200127, P. R. China
| | - Xiaochen Tang
- Clinical Laboratory, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Shanghai Key Laboratory of Clinical Molecular Diagnostics for Pediatrics, Shanghai, 200127, P. R. China
| | - Rui Zhu
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, 200072, P. R. China
| | - Dongsheng Mao
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, 200072, P. R. China
| | - Xiaoli Zhu
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai, 200072, P. R. China
| | - Qiuhui Pan
- Clinical Laboratory, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Shanghai Key Laboratory of Clinical Molecular Diagnostics for Pediatrics, Shanghai, 200127, P. R. China
- Sanya Women and Children's Hospital Managed by Shanghai Children's Medical Center, Sanya, 572000, P. R. China
| |
Collapse
|
93
|
Peeters G, Verhaegen A. Severe insulin resistance in a patient with diabetes after treatment with brentuximab vedotin. BMJ Case Rep 2024; 17:e251867. [PMID: 38569738 PMCID: PMC10989180 DOI: 10.1136/bcr-2022-251867] [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] [Indexed: 04/05/2024] Open
Abstract
A man in his late 60s with a history of well-controlled type 2 diabetes and hepatic cirrhosis presented to the emergency department due to uncontrollable hyperglycaemia following the initial brentuximab vedotin (BV) infusion. BV was initiated as a treatment for mycosis fungoides, a form of cutaneous T-cell lymphoma. The patient was diagnosed with severe hyperglycaemia with ketosis. Empiric treatment with amoxicillin-clavulanic acid, hydration and intravenous insulin infusion was initiated. Hyperglycaemia persisted despite receiving massive amounts of insulin and was corrected only after treatment with high-dose methylprednisolone for suspected type B insulin resistance. Extremely high and difficult-to-treat hyperglycaemia is a rare side effect of BV. Unfortunately, the patient died of upper gastrointestinal bleeding 22 days after discharge. In patients with obesity and/or diabetes mellitus, the blood glucose levels should be carefully monitored when treated with BV.
Collapse
Affiliation(s)
- Greet Peeters
- Endocrinology-Diabetology, ZNA Jan Palfijn, Merksem, Belgium
| | - Ann Verhaegen
- Endocrinology-Diabetology, ZNA Jan Palfijn, Merksem, Belgium
| |
Collapse
|
94
|
Liu S, Liu F, Hou X, Zhang Q, Ren Y, Zhu H, Yang Z, Xu X. KRAS Mutation Detection with (2 S,4 R)-4-[ 18F]FGln for Noninvasive PDAC Diagnosis. Mol Pharm 2024; 21:2034-2042. [PMID: 38456403 PMCID: PMC10989612 DOI: 10.1021/acs.molpharmaceut.4c00082] [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: 01/25/2024] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/09/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), which has a poor prognosis and nonspecific symptoms and progresses rapidly, is the most common pancreatic cancer type. Inhibitors targeting KRAS G12D and G12C mutations have been pivotal in PDAC treatment. Cancer cells with different KRAS mutations exhibit various degrees of glutamine dependency; in particular, cells with KRAS G12D mutations exhibit increased glutamine uptake. (2S,4R)-4-[18F]FGln has recently been developed for clinical cancer diagnosis and tumor cell metabolism analysis. Thus, we verified the heterogeneity of glutamine dependency in PDAC models with different KRAS mutations by a visual and noninvasive method with (2S,4R)-4-[18F]FGln. Two tumor-bearing mouse models (bearing the KRAS G12D or G12C mutation) were injected with (2S,4R)-4-[18F]FGln, and positron emission tomography (PET) imaging features and biodistribution were observed and analyzed. The SUVmax in the regions of interest (ROI) was significantly higher in PANC-1 (G12D) tumors than in MIA PaCa-2 (G12C) tumors. Biodistribution analysis revealed higher tumor accumulation of (2S,4R)-4-[18F]FGln and other metrics, such as T/M and T/B, in the PANC-1 mouse models compared to those in the MIAPaCa-2 mouse models. In conclusion, PDAC cells with the KRAS G12D and G12C mutations exhibit various degrees of (2S,4R)-4-[18F]FGln uptake, indicating that (2S,4R)-4-[18F]FGln might be applied to detect KRAS G12C and G12D mutations and provide treatment guidance.
Collapse
Affiliation(s)
| | | | - Xingguo Hou
- State Key Laboratory
of Holistic
Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory
of Carcinogenesis and Translational Research, NMPA Key Laboratory
for Research and Evaluation of Radiopharmaceuticals (National Medical
Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Qian Zhang
- State Key Laboratory
of Holistic
Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory
of Carcinogenesis and Translational Research, NMPA Key Laboratory
for Research and Evaluation of Radiopharmaceuticals (National Medical
Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ya’nan Ren
- State Key Laboratory
of Holistic
Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory
of Carcinogenesis and Translational Research, NMPA Key Laboratory
for Research and Evaluation of Radiopharmaceuticals (National Medical
Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Hua Zhu
- State Key Laboratory
of Holistic
Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory
of Carcinogenesis and Translational Research, NMPA Key Laboratory
for Research and Evaluation of Radiopharmaceuticals (National Medical
Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhi Yang
- State Key Laboratory
of Holistic
Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory
of Carcinogenesis and Translational Research, NMPA Key Laboratory
for Research and Evaluation of Radiopharmaceuticals (National Medical
Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiaoxia Xu
- State Key Laboratory
of Holistic
Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory
of Carcinogenesis and Translational Research, NMPA Key Laboratory
for Research and Evaluation of Radiopharmaceuticals (National Medical
Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
95
|
Banesh S, Patil N, Chethireddy VR, Bhukmaria A, Saudagar P. Design and evaluation of a multiepitope vaccine for pancreatic cancer using immune-dominant epitopes derived from the signature proteome in expression datasets. Med Oncol 2024; 41:90. [PMID: 38522058 DOI: 10.1007/s12032-024-02334-4] [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: 11/06/2023] [Accepted: 02/14/2024] [Indexed: 03/25/2024]
Abstract
Pancreatic cancer is a highly aggressive and often lethal malignancy with limited treatment options. Its late-stage diagnosis and resistance to conventional therapies make it a significant challenge in oncology. Immunotherapy, particularly cancer vaccines, has emerged as a promising avenue for treating pancreatic cancer. Multi-epitope vaccines, designed to target multiple epitopes derived from various antigens associated with pancreatic cancer, have gained attention as potential candidates for improving therapeutic outcomes. In this study, we have explored transcriptomics and protein expression databases to identify potential upregulated proteins in pancreatic cancer cells. After examining a total of 21,054 proteins from various databases, it was discovered that 143 proteins expressed differently in malignant and healthy cells. The CTL, HTL and BCE epitopes were predicted for the shortlisted proteins. 51,840 vaccine constructs were created by concatenating CTL, HTL, and B-cell epitopes in the respective sequences. The best 86 structures were selected from a set of 51,840 designs after they were analyzed for vaxijenicity, allergenicity, toxicity, and antigenicity scores. In further simulation of the immune response using constructs, it was found that 41417, 37961, and 40841 constructs could produce a strong immune response when injected. Further, it was found that construct 37961 showed stronger interaction and stability with TLR-9 as determined from the large-scale molecular dynamics simulations. Moreover, the 37961 construct has shown interactions with TLR-9 suggests its potential in inducing immune response. In addition, construct 37961 has shown 100% predicted solubility in the E. coli expression system. Overall, the study indicates the designed construct 37961 has the potential to induce an anti-tumor immune response and long-standing protection pending further studies.
Collapse
Affiliation(s)
- Sooram Banesh
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal, Telangana, 506004, India
| | - Nupoor Patil
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal, Telangana, 506004, India
| | - Vihadhar Reddy Chethireddy
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal, Telangana, 506004, India
| | - Arnav Bhukmaria
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal, Telangana, 506004, India
| | - Prakash Saudagar
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal, Telangana, 506004, India.
| |
Collapse
|
96
|
Wu H, Zhang XH, Wang LP, Tian HD, Liu GR, Yang DH, Liu SL. Successful Outcome of a Patient with Concomitant Pancreatic and Renal Carcinoma Receiving Secoisolariciresinol Diglucoside Therapy Alone: A Case Report. Int Med Case Rep J 2024; 17:167-175. [PMID: 38504721 PMCID: PMC10949998 DOI: 10.2147/imcrj.s446184] [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: 10/23/2023] [Accepted: 03/06/2024] [Indexed: 03/21/2024] Open
Abstract
Introduction Pancreatic cancer (PC) is among the deadliest malignancies. Kidney cancer (KC) is a common malignancy globally. Chemo- or radio-therapies are not very effective to control PC or KC, and overdoses often cause severe site reactions to the patients. As a result, novel treatment strategies with high efficacy but without toxic side effects are urgently desired. Secoisolariciresinol diglucoside (SDG) belongs to plant lignans with potential anticancer activities, but clinical evidence is not available in PC or KC treatment. Patient Concerns We report a rare case of an 83-year-old female patient with pancreatic and kidney occupying lesions that lacked the conditions to receive surgery or chemo- or radiotherapy. Diagnosis Pancreatic and kidney cancers. Interventions We gave dietary SDG to the patient as the only therapeutics. Outcomes SDG effectively halted progression of both PC and KC. All clinical manifestations, including bad insomnia, loss of appetite, stomach symptoms, and skin itching over the whole body, all disappeared. The initial massive macroscopic hematuria became microscopic and infrequent, and other laboratory results also gradually returned to normal. Most of the cancer biomarkers, initially high such as CEA, CA199, CA724, CA125, came down rapidly, among which CA199 changed most radically. This patient has had progression-free survival of one year so far. Conclusion These results demonstrate the potent inhibitory effects of SDG on PC and KC of this patient and provide promising novel therapeutics for refractory malignant tumors.
Collapse
Affiliation(s)
- Hao Wu
- Genomics Research Center (State-Province Key Laboratory of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, Harbin, People’s Republic of China
- Key Laboratory of Tumor Biotherapy of Heilongjiang Province, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
- Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, People’s Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, People’s Republic of China
- Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, People’s Republic of China
- Translational Medicine Research and Cooperation Center of Northern China, Harbin Medical University, Harbin, People’s Republic of China
| | - Xing-Hua Zhang
- Genomics Research Center (State-Province Key Laboratory of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, Harbin, People’s Republic of China
- Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, People’s Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, People’s Republic of China
- Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, People’s Republic of China
- Translational Medicine Research and Cooperation Center of Northern China, Harbin Medical University, Harbin, People’s Republic of China
| | - Li-Ping Wang
- KangYuan Hospital, Harbin, People’s Republic of China
- Xun-Qi Medicine Clinic, Harbin, People’s Republic of China
| | - Hong-Da Tian
- Genomics Research Center (State-Province Key Laboratory of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, Harbin, People’s Republic of China
- Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, People’s Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, People’s Republic of China
- Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, People’s Republic of China
- Translational Medicine Research and Cooperation Center of Northern China, Harbin Medical University, Harbin, People’s Republic of China
| | - Gui-Rong Liu
- Genomics Research Center (State-Province Key Laboratory of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, Harbin, People’s Republic of China
- Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, People’s Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, People’s Republic of China
- Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, People’s Republic of China
- Translational Medicine Research and Cooperation Center of Northern China, Harbin Medical University, Harbin, People’s Republic of China
| | - Dong-Hui Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, People’s Republic of China
| | - Shu-Lin Liu
- Genomics Research Center (State-Province Key Laboratory of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, Harbin, People’s Republic of China
- Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, People’s Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, People’s Republic of China
- Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, People’s Republic of China
- Translational Medicine Research and Cooperation Center of Northern China, Harbin Medical University, Harbin, People’s Republic of China
- KangYuan Hospital, Harbin, People’s Republic of China
- Xun-Qi Medicine Clinic, Harbin, People’s Republic of China
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Canada
| |
Collapse
|
97
|
Xia DQ, Zhou Y, Yang S, Li FF, Tian LY, Li YH, Xu HY, Xiao CZ, Wang W. Combining prognostic value of serum carbohydrate antigen 19-9 and tumor size reduction ratio in pancreatic ductal adenocarcinoma. World J Gastrointest Oncol 2024; 16:798-809. [PMID: 38577439 PMCID: PMC10989379 DOI: 10.4251/wjgo.v16.i3.798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/15/2023] [Accepted: 01/27/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a common cancer with increasing morbidity and mortality due to changes of social environment. AIM To evaluate the significance of serum carbohydrate antigen 19-9 (CA19-9) and tumor size changes pre- and post-neoadjuvant therapy (NAT). METHODS This retrospective study was conducted at the Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital. This study specifically assessed CA19-9 levels and tumor size before and after NAT. RESULTS A total of 156 patients who completed NAT and subsequently underwent tumor resection were included in this study. The average age was 65.4 ± 10.6 years and 72 (46.2%) patients were female. Before survival analysis, we defined the post-NAT serum CA19-9 level/pre-NAT serum CA19-9 level as the CA19-9 ratio (CR). The patients were divided into three groups: CR < 0.5, CR > 0.5 and < 1 and CR > 1. With regard to tumor size measured by both computed tomography and magnetic resonance imaging, we defined the post-NAT tumor size/pre-NAT tumor size as the tumor size ratio (TR). The patients were then divided into three groups: TR < 0.5, TR > 0.5 and < 1 and TR > 1. Based on these groups divided according to CR and TR, we performed both overall survival (OS) and disease-free survival (DFS) analyses. Log-rank tests showed that both OS and DFS were significantly different among the groups according to CR and TR (P < 0.05). CR and TR after NAT were associated with increased odds of achieving a complete or near-complete pathologic response. Moreover, CR (hazard ratio: 1.721, 95%CI: 1.373-3.762; P = 0.006), and TR (hazard ratio: 1.435, 95%CI: 1.275-4.363; P = 0.014) were identified as independent factors associated with OS. CONCLUSION This study demonstrated that post-NAT serum CA19-9 level/pre-NAT serum CA19-9 level and post-NAT tumor size/pre-NAT tumor size were independent factors associated with OS in patients with PDAC who received NAT and subsequent surgical resection.
Collapse
Affiliation(s)
- Dong-Qin Xia
- Oncology Treatment Center of Traditional Chinese Medicine, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yong Zhou
- Department of Oncology, Chongqing Weisiteng Biotech Translational Research Institute, Chongqing 430039, China
- Chongqing Key Laboratory on Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing 430065, China
| | - Shuang Yang
- Oncology Treatment Center of Traditional Chinese Medicine, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Fang-Fei Li
- Oncology Treatment Center of Traditional Chinese Medicine, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Li-Ya Tian
- Oncology Treatment Center of Traditional Chinese Medicine, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yan-Hua Li
- Oncology Treatment Center of Traditional Chinese Medicine, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Hai-Yan Xu
- Oncology Treatment Center of Traditional Chinese Medicine, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Cai-Zhi Xiao
- Oncology Treatment Center of Traditional Chinese Medicine, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Wei Wang
- Oncology Treatment Center of Traditional Chinese Medicine, Chongqing University Cancer Hospital, Chongqing 400030, China
| |
Collapse
|
98
|
Xiao C, Li J, Hua A, Wang X, Li S, Li Z, Xu C, Zhang Z, Yang X, Li Z. Hyperbaric Oxygen Boosts Antitumor Efficacy of Copper-Diethyldithiocarbamate Nanoparticles against Pancreatic Ductal Adenocarcinoma by Regulating Cancer Stem Cell Metabolism. RESEARCH (WASHINGTON, D.C.) 2024; 7:0335. [PMID: 38766644 PMCID: PMC11100349 DOI: 10.34133/research.0335] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 02/16/2024] [Indexed: 05/22/2024]
Abstract
Cuproptosis-based cancer nanomedicine has received widespread attention recently. However, cuproptosis nanomedicine against pancreatic ductal adenocarcinoma (PDAC) is severely limited by cancer stem cells (CSCs), which reside in the hypoxic stroma and adopt glycolysis metabolism accordingly to resist cuproptosis-induced mitochondria damage. Here, we leverage hyperbaric oxygen (HBO) to regulate CSC metabolism by overcoming tumor hypoxia and to augment CSC elimination efficacy of polydopamine and hydroxyethyl starch stabilized copper-diethyldithiocarbamate nanoparticles (CuET@PH NPs). Mechanistically, while HBO and CuET@PH NPs inhibit glycolysis and oxidative phosphorylation, respectively, the combination of HBO and CuET@PH NPs potently suppresses energy metabolism of CSCs, thereby achieving robust tumor inhibition of PDAC and elongating mice survival importantly. This study reveals novel insights into the effects of cuproptosis nanomedicine on PDAC CSC metabolism and suggests that the combination of HBO with cuproptosis nanomedicine holds significant clinical translation potential for PDAC patients.
Collapse
Affiliation(s)
- Chen Xiao
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Jiayuan Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Ao Hua
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Xing Wang
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Shiyou Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Zheng Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Chen Xu
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Zhijie Zhang
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Xiangliang Yang
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- National Engineering Research Center for Nanomedicine,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Key Laboratory of Molecular Biophysics of Ministry of Education,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Hubei Bioinformatics and Molecular Imaging Key Laboratory,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Zifu Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- National Engineering Research Center for Nanomedicine,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Key Laboratory of Molecular Biophysics of Ministry of Education,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Hubei Bioinformatics and Molecular Imaging Key Laboratory,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| |
Collapse
|
99
|
Xu Q, Jia C, Ou Y, Zeng C, Jia Y. Dark horse target Claudin18.2 opens new battlefield for pancreatic cancer. Front Oncol 2024; 14:1371421. [PMID: 38511141 PMCID: PMC10951399 DOI: 10.3389/fonc.2024.1371421] [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: 01/16/2024] [Accepted: 02/23/2024] [Indexed: 03/22/2024] Open
Abstract
Pancreatic cancer is one of the deadliest malignant tumors, which is a serious threat to human health and life, and it is expected that pancreatic cancer may be the second leading cause of cancer death in developed countries by 2030. Claudin18.2 is a tight junction protein expressed in normal gastric mucosal tissues, which is involved in the formation of tight junctions between cells and affects the permeability of paracellular cells. Claudin18.2 is highly expressed in pancreatic cancer and is associated with the initiation, progression, metastasis and prognosis of cancer, so it is considered a potential therapeutic target. Up to now, a number of clinical trials for Claudin18.2 are underway, including solid tumors such as pancreatic cancers and gastric cancers, and the results of these trials have not yet been officially announced. This manuscript briefly describes the Claudia protein, the dual roles of Cluadin18 in cancers, and summarizes the ongoing clinical trials targeting Claudin18.2 with a view to integrating the research progress of Claudin18.2 targeted therapy. In addition, this manuscript introduces the clinical research progress of Claudin18.2 positive pancreatic cancer, including monoclonal antibodies, bispecific antibodies, antibody-drug conjugates, CAR-T cell therapy, and hope to provide feasible ideas for the clinical treatment of Claudin18.2 positive pancreatic cancer.
Collapse
Affiliation(s)
- Qian Xu
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Department of Oncology, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Caiyan Jia
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Department of Oncology, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yan Ou
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Department of Oncology, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Chuanxiu Zeng
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Department of Oncology, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yingjie Jia
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Department of Oncology, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
100
|
Wang L, Wang G, Wang P, Nie F. Pancreatic ductal adenocarcinoma: CEUS characteristics are correlated with pathological findings and help predict early recurrence after resection. JOURNAL OF CLINICAL ULTRASOUND : JCU 2024; 52:230-240. [PMID: 38018362 DOI: 10.1002/jcu.23622] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/30/2023]
Abstract
OBJECTIVES To identify characteristics of preoperative contrast-enhanced ultrasound (CEUS) that could predict early recurrence after curative resection of pancreatic ductal adenocarcinoma (PDAC). METHODS From January 2017 to September 2022, a total of 110 patients with PDAC (all confirmed by samples obtained via operation) who underwent CEUS within 1 month before surgery were enrolled. We proposed five CEUS enhancement patterns (Pattern I, homogeneous enhancement; Pattern II, heterogeneous enhancement without cystic components; pattern III, ring enhancement; Pattern IV, starry enhancement; Pattern V, heterogeneous enhancement with cystic components) of PDAC. Clinical-pathologic and CEUS characteristics for predicting early recurrence (recurrence within 1 year after curative resection) were analyzed. Important CEUS characteristics were compared with the pathological findings. RESULTS Tumor size and TNM stage were closely associated with early recurrence. Incomplete-enhancement and enhancement pattern III, IV and V at CEUS imaging were more prone to early recurrence. Incomplete-enhancement lesions had higher histological tumor grades, less frequent remaining acini, and more frequent necrosis within the tumor. PDACs with pattern I and II had lower histological tumor grades, and pattern III, IV and V had higher histological tumor grades. PDACs with pattern I, II and IV had less frequent intratumoral necrosis than PDACs with pattern III and V, and PDACs with pattern IV had lower MVD values. CONCLUSIONS PDACs with incomplete enhancement and enhancement pattern III, IV and V were more prone to early recurrence after attempted curative resection, and these important CEUS characteristics were closely related to the pathological findings of PDAC.
Collapse
Affiliation(s)
- Lan Wang
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Guojuan Wang
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Peihua Wang
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Fang Nie
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|