1
|
Sugawara S, Sone M, Umino R, Ban D, Itou C, Kimura S, Oshima T, Ozawa M, Tanishima T, Nakama R, Murakami S, Kusumoto M, Mizui T, Takamoto T, Nara S, Esaki M. Safety and feasibility of percutaneous abdominal lavage cytology screening (PACS) prior to surgical resection for pancreatic cancer. Abdom Radiol (NY) 2024; 49:4365-4372. [PMID: 39120716 DOI: 10.1007/s00261-024-04510-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 08/10/2024]
Abstract
PURPOSE This study aimed to evaluate the feasibility, safety, diagnostic yield, and technical aspects of percutaneous abdominal lavage cytology screening (PACS) in patients with resectable pancreatic cancer. METHODS This single-center, retrospective study included patients with resectable pancreatic cancer who underwent PACS before pancreatectomy between May 2022 and October 2023. The technical success rate, position of the drainage tube, volume of fluid administered, volume of fluid retrieved, fluid retrieval rate, and adverse events were evaluated. The cytological results of PACS were compared with those of surgical peritoneal lavage performed during pancreatectomy. RESULTS Forty-four patients were enrolled in this study. The technical success rate for PACS was 100%. Drainage tube placement was outside the pouch of Douglas in all patients in the right-sided abdominal approach group (n = 10), whereas the placement was in the pouch of Douglas in all patients in the suprapubic approach group (n = 34). The mean volume of fluid administered, mean volume of fluid retrieved, and fluid retrieval rate were 185.0 ± 22.9 ml vs. 97.1 ± 32.0 ml (p < 0.001), 36.8 ± 25.6 ml vs. 50.5 ± 21.6 ml (p = 0.059), and 19.0 ± 12.4% vs. 54.9 ± 21.9% (p < 0.001) in the right abdominal approach and suprapubic approach groups, respectively. No adverse events were reported. The cytological results were benign in 42 patients; no discrepancy was observed in the results of surgical peritoneal lavage (n = 36). CONCLUSION PACS is a feasible and safe procedure that can be performed before pancreatectomy in patients with resectable pancreatic cancer. the suprapubic approach may be ideal and PACS could be a screening method to detect carcinomatous peritonitis.
Collapse
Affiliation(s)
- Shunsuke Sugawara
- Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| | - Miyuki Sone
- Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Ryosuke Umino
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Daisuke Ban
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Chihiro Itou
- Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Shintaro Kimura
- Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Takumi Oshima
- Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Mizuki Ozawa
- Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Tomoya Tanishima
- Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Rakuhei Nakama
- Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Sho Murakami
- Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Masahiko Kusumoto
- Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Takahiro Mizui
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Takeshi Takamoto
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Satoshi Nara
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Minoru Esaki
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| |
Collapse
|
2
|
Yang W, Hu P, Zuo C. Application of imaging technology for the diagnosis of malignancy in the pancreaticobiliary duodenal junction (Review). Oncol Lett 2024; 28:596. [PMID: 39430731 PMCID: PMC11487531 DOI: 10.3892/ol.2024.14729] [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: 04/15/2024] [Accepted: 09/13/2024] [Indexed: 10/22/2024] Open
Abstract
The pancreaticobiliary duodenal junction (PBDJ) is the connecting area of the pancreatic duct, bile duct and duodenum. In a broad sense, it refers to a region formed by the head of the pancreas, the pancreatic segment of the common bile duct and the intraduodenal segment, the descending and the horizontal part of the duodenum, and the soft tissue around the pancreatic head. In a narrow sense, it refers to the anatomical Vater ampulla. Due to its complex and variable anatomical features, and the diversity of pathological changes, it is challenging to make an early diagnosis of malignancy at the PBDJ and define the histological type. The unique anatomical structure of this area may be the basis for the occurrence of malignant tumors. Therefore, understanding and subclassifying the anatomical configuration of the PBDJ is of great significance for the prevention and treatment of malignant tumors at their source. The present review comprehensively discusses commonly used imaging techniques and other new technologies for diagnosing malignancy at the PBDJ, offering evidence for physicians and patients to select appropriate examination methods.
Collapse
Affiliation(s)
- Wanyi Yang
- Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Clinical Research Center for Tumor of Pancreaticobiliary Duodenal Junction in Hunan Province, Changsha, Hunan 410013, P.R. China
- Graduates Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Changsha, Hunan 410013, P.R. China
| | - Pingsheng Hu
- Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Clinical Research Center for Tumor of Pancreaticobiliary Duodenal Junction in Hunan Province, Changsha, Hunan 410013, P.R. China
| | - Chaohui Zuo
- Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Clinical Research Center for Tumor of Pancreaticobiliary Duodenal Junction in Hunan Province, Changsha, Hunan 410013, P.R. China
- Graduates Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
3
|
Chang S, Zou Y, Huang J, Li Z, Liang Y, Gao S. Fibrinogen to pre-albumin ratio is an independent prognostic index for patients with pancreatic ductal adenocarcinoma after radical resection. World J Surg Oncol 2024; 22:284. [PMID: 39468569 PMCID: PMC11520588 DOI: 10.1186/s12957-024-03524-0] [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: 05/10/2024] [Accepted: 09/01/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND This study aims to elucidate the significance of the preoperative fibrinogen to pre-albumin ratio (FPR) in predicting the prognosis of pancreatic ductal adenocarcinoma (PDAC), a correlation not extensively explored previously. METHODS A cohort of 563 patients diagnosed with PDAC and subjected to radical surgical resection was examined. We meticulously documented a range of inflammatory markers, clinical-pathological features, and oncological outcomes. The prognostic value of preoperative FPR was assessed using Kaplan-Meier survival analysis and Cox proportional hazards regression modeling. Furthermore, the predictive accuracy of FPR was evaluated through time-dependent receiver operating characteristic (ROC) curves and decision curve analyses (DCA). RESULTS The determined optimal threshold for FPR was 14.77, which facilitated the stratification of patients into groups with low and high FPR levels. Notably, patients in the high FPR cohort exhibited significantly reduced recurrence-free survival (RFS) and overall survival (OS) rates compared to their low FPR counterparts. Multivariate Cox regression analysis underscored FPR as an independent prognostic indicator for both RFS and OS. In comparison to the neutrophil-to-lymphocyte ratio (NLR), FPR demonstrated superior prognostic accuracy and clinical utility. CONCLUSION The preoperative fibrinogen to pre-albumin ratio serves as an independent prognostic marker for RFS and OS among PDAC patients undergoing radical resection. Our findings suggest that FPR could be a valuable addition to the current prognostic models, potentially guiding therapeutic decision-making and patient management strategies in PDAC.
Collapse
Affiliation(s)
- Shaofei Chang
- Department of General Surgery, Shanxi Provincial People's Hospital, Taiyuan, PR, China
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Yiping Zou
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Jing Huang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Zhifei Li
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Yuexiang Liang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China.
- Department of Gastrointestinal Oncology, the first affiliated hospital of , Hainan Medical University, Haikou, PR, China.
| | - Song Gao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China.
| |
Collapse
|
4
|
Wang N, Chai T, Wang XR, Zheng YD, Sang CY, Yang JL. Pin1: Advances in pancreatic cancer therapeutic potential and inhibitors research. Bioorg Chem 2024; 153:107869. [PMID: 39418844 DOI: 10.1016/j.bioorg.2024.107869] [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: 05/29/2024] [Revised: 08/18/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024]
Abstract
The peptidyl-prolyl cis/trans isomerase NIMA-interaction 1 (Pin1) catalyzes the transition of the proline ring from the cis to trans conformation, resulting in conformational and functional changes in proteins that are regulated by proline-guided serine/threonine phosphorylation. In recent years, Pin1 has emerged as a novel molecular target for the diagnosis and treatment of various malignant tumors. Notably, it has been found that Pin1 is highly expressed in pancreatic cancer. This article focuses on the mechanisms by which Pin1 orchestrates multiple oncogenic functions in the development of pancreatic cancer. By exploring the intricate interactions between Pin1 and the pancreatic tumor microenvironment, we provide an overview of Pin1's role in modifying glycolytic metabolism, redox balance, and the hypoxic microenvironment of pancreatic cancer. Furthermore, we summarize the potential anticancer effects of Pin1 inhibitors, aiming to elucidate Pin1's promise as a potential anticancer agent, particularly in the context of pancreatic cancer.
Collapse
Affiliation(s)
- Nan Wang
- College of Pharmacy, Gansu University of Chinese Medicine; CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, China
| | - Tian Chai
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, China
| | - Xing-Rong Wang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, China
| | - Yi-Dan Zheng
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, China
| | - Chun-Yan Sang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, China
| | - Jun-Li Yang
- College of Pharmacy, Gansu University of Chinese Medicine; CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, China.
| |
Collapse
|
5
|
Jia ZY, Yan X, Zhou H, Wang W, Li C, Zhang BL. Anti-PD-L1 checkpoint inhibitor combined with nanocarrier-mediated cisplatin codelivery system for effective treatment of pancreatic cancer. Mol Immunol 2024; 174:69-76. [PMID: 39216237 DOI: 10.1016/j.molimm.2024.08.006] [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: 06/14/2024] [Revised: 07/30/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
Immune checkpoint inhibitor-based cancer immunotherapy has shown promise as a potential treatment in the clinic. It has been reported that anti-PD-L1 combined with cisplatin treatment can improve the antitumor effect. However, the therapeutic outcome is limited due to the abundance of tumor stroma in pancreatic cancer (PC), which prevented the penetration of cisplatin and anti-PD-L1 into tumor regions, thus impeding the effectiveness in the treatment of PC. In this study, a nanocarrier-mediated codelivery system of hyaluronidase and cisplatin was constructed, which can degrade the stroma and promote cisplatin and anti-PD-L1 to penetrate the tumor stroma into the deep tumor, so as to suppress PC effectively. When combined the cisplatin nanocarrier system BPEI-SS-Pt/HAase@CaP (BSP/H@CaP) with an immune checkpoint inhibitor to overcome the poor therapeutic outcome of PC, the results indicated that the therapeutic effect of BSP/H@CaP combined with anti-PD-L1 was better than that of BSP/H@CaP and single anti-PD-L1 group. Because the stroma is degrading, a higher amount of BPEI-SS-Pt and anti-PD-L1 can enter the tumor stroma and reach the inner depths of the tumor for immune stimulation, leading to a synergistically augmented chemotherapy and immunotherapy for PC. The above combination therapy is useful for clinical translation to overcome the treatment resistance of matrix-rich PC.
Collapse
Affiliation(s)
- Zhou-Yan Jia
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, and Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine, Xi'an 710032, China; School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xin Yan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, School of Stomatology Oral and Maxillofacial Surgery, Fourth Military Medical University, Xi'an 71000, China
| | - Hao Zhou
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, and Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine, Xi'an 710032, China
| | - Wei Wang
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, and Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine, Xi'an 710032, China.
| | - Chen Li
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, and Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine, Xi'an 710032, China
| | - Bang-Le Zhang
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, and Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine, Xi'an 710032, China.
| |
Collapse
|
6
|
Chen Y, Ma C, Yang P, Mao K, Gao Y, Chen L, Wang Z, Bian Y, Shao C, Lu J. Values of apparent diffusion coefficient in pancreatic cancer patients receiving neoadjuvant therapy. BMC Cancer 2024; 24:1160. [PMID: 39294623 PMCID: PMC11412028 DOI: 10.1186/s12885-024-12934-y] [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] [Accepted: 09/11/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND To investigate the values of apparent diffusion coefficient (ADC) for the treatment response evaluation in pancreatic cancer (PC) patients receiving neoadjuvant therapy (NAT). METHODS This study included 103 NAT patients with histologically proven PC. ADC maps were generated using monoexponential diffusion-weighted imaging (b values: 50, 800 s/mm2). Tumors' minimum, maximum, and mean ADCs were measured and compared pre- and post-NAT. Variations in ADC values measured between pre- and post-NAT completion for NAT methods (chemotherapy, chemoradiotherapy), tumor locations (head/neck, body/tail), tumor regression grade (TRG) levels (0-2, 3), N stages (N0, N1/N2) and tumor resection margin status (R0, R1), were further analyzed. RESULTS The minimum, maximum, and mean ADC values all increased dramatically after NAT, rising from 23.4 to 25.4% (all p < 0.001): mean (average: 1.626 × 10- 3 mm2/s vs. 1.315 × 10- 3 mm2/s), minimum (median: 1.274 × 10- 3 mm2/s vs. 1.034 × 10- 3 mm2/s), and maximum (average: 1.981 × 10- 3 mm2/s vs. 1.580 × 10- 3 mm2/s). The ADCs between the subgroups of all the criteria under investigation did not differ significantly for the minimum, maximum, or mean values pre- or post-NAT (P = 0.08 to 1.00). In the patients with borderline resectable PC (n = 47), the rate of tumor size changes after NAT was correlated with the pre-NAT mean ADC values (Spearman's coefficient: 0.288, P = 0.049). CONCLUSIONS The ADC values of PC increased significantly following NAT; however, the percentage increases failed to provide any predictive value for the resection margin status or TRG levels.
Collapse
Affiliation(s)
- Yufei Chen
- College of Electronic and Information Engineering, Tongji University, Shanghai, China
| | - Chao Ma
- College of Electronic and Information Engineering, Tongji University, Shanghai, China.
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Changhai Road 168, Shanghai, 200434, China.
| | - Panpan Yang
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Kuanzheng Mao
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yisha Gao
- Department of Pathology, Changhai Hospital of Shanghai, Naval Medical University, Shanghai, China
| | - Luguang Chen
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Zhen Wang
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Yun Bian
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Chengwei Shao
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| | - Jianping Lu
- Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, Changhai Road 168, Shanghai, 200434, China
| |
Collapse
|
7
|
Zhang H, Liu A, Bo W, Zhang M, Wang H, Feng X, Wu Y. Upregulation of HSD11B1 promotes cortisol production and inhibits NK cell activation in pancreatic adenocarcinoma. Mol Immunol 2024; 175:10-19. [PMID: 39276709 DOI: 10.1016/j.molimm.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 08/12/2024] [Accepted: 08/22/2024] [Indexed: 09/17/2024]
Abstract
Cortisol is a glucocorticoid hormone that has immunosuppressive function. Elevated basal cortisol levels are present in patients with some kinds of cancers, but its role in the microenvironment of pancreatic adenocarcinoma (PAAD) remains unclear. This study analyzed the expression of genes involved in cortisol generation by using high-throughput sequencing data from TCGA portal and found HSD11B1 was significantly upregulated in patients with PAAD. The correlations between HSD11B1 level and the expression of 23 immunosuppressive receptors were analyzed by Spearman's correlation analysis. The function of HSD11B1 was examined in primary NK cells and PAAD cell lines. The levels of cortisol in medium and cell lysates were detected by ELISA. In vitro killing assay was used to evaluate the cytotoxicity of NK cells. Cell surface levels of CD96, Tim-3, PD-1, TIGIT, CTLA-4, NKp46, NKp30, NKD2G and LFA-1A, and intracellular levels of CD107a and IFN-γ were examined by flow cytometry. We observed that patients with higher HSD11B1 level had shorter survival time. HSD11B1 is positively correlated with the mRNA levels of 11 immunosuppressive receptors in PAAD. Higher HSD11B1 level relates to reduced abundance of activated NK cells in the tumors. HSD11B1 overexpressed NK cells exhibit exhausted phenotype with increased cortisol production, reduced viability, and reduced cytotoxicity against cancer cells. Overexpression of HSD11B1 did not change the viability of tumor cells but upregulated cortisol production. Targeting HSD11B1 by a specific inhibitor improved the NK cells responsiveness. In conclusion, HSD11B1 is upregulated in patients with PAAD, and higher HSD11B1 level is related to poor prognosis. Upregulation of HSD11B1 in NK and tumor cells increased the production and secretion of cortisol and induces NK cell exhaustion.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Aixiang Liu
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Wentao Bo
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Mingyi Zhang
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Haiqing Wang
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Xielin Feng
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Ying Wu
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China; Department of Medical Oncology, Daytime Medical Treatment Area, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
8
|
Li MM, Zhang Y, Sun F, Huai MX, Zhang FY, Pan JX, Qu CY, Shen F, Li ZH, Xu LM. Photodynamic Therapy Using RGD-Functionalized Quantum Dots Elicit a Potent Immune Response in a Syngeneic Mouse Model of Pancreatic Cancer. Int J Nanomedicine 2024; 19:9487-9502. [PMID: 39290860 PMCID: PMC11406538 DOI: 10.2147/ijn.s479123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024] Open
Abstract
Purpose Photodynamic therapy (PDT) induces anti-tumor immune responses by triggering immunogenic cell death in tumor cells. Previously, we demonstrated that novel QDs-RGD nanoparticles exhibited high efficiency as photosensitizers in the treatment of pancreatic cancer. However, the underlying mechanism of the anti-tumor immune effects induced by the photosensitizer remains unknown. This study assessed the anticancer immune effect of QDs-RGD, as well as the conventional photosensitizer chlorine derivative, YLG-1, for comparison, against pancreatic cancer in support of superior therapeutic efficacy. Methods The pancreatic cancer cell line, Panc02, was used for in vitro studies. C57BL/6 mice bearing pancreatic cancer cell-derived xenografts were generated for in vivo studies to assess the anti-tumor effects of QDs-RGD-PDT and YLG-1-PDT. The immunostimulatory ability of both photosensitizers was examined by measuring the expression of damage-associated molecular patterns (DAMP), such as calreticulin (CRT), assessing dendritic cell (DC) maturation, and analyzing cytokine expression. The specific immunity of QDs-RGD and YLG-1-PDT on distant tumor were determined by combining PDT with anti-CTLA-4 antibody. Results QDs-RGD-PDT and YLG-1-PDT significantly inhibited pancreatic cancer cell growth in a dose- and time-dependent manner. While both photosensitizers significantly promoted CRT release, DC maturation, and interferon γ (IFN-γ) and tumor necrosis factor α (TNF-α) expression, QDs-RGD exerted a stronger immunostimulatory effect than YLG-1. Combination treatment with QDs-RGD and CTLA-4 blockade was able to significantly inhibit the growth of distant tumors. Conclusion QDs-RGD is a novel and effective PDT strategy for treating pancreatic tumors by inducing anti-tumor immune responses.
Collapse
Affiliation(s)
- Ming-Ming Li
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Yi Zhang
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Fang Sun
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Man-Xiu Huai
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Fei-Yu Zhang
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Jia-Xing Pan
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Chun-Ying Qu
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Feng Shen
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Zheng-Hong Li
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Lei-Ming Xu
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| |
Collapse
|
9
|
Boggi U, Kauffmann EF, Napoli N, Barreto SG, Besselink MG, Fusai GK, Hackert T, Hilal MA, Marchegiani G, Salvia R, Shrikhande SV, Truty M, Werner J, Wolfgang C, Bannone E, Capretti G, Cattelani A, Coppola A, Cucchetti A, De Sio D, Di Dato A, Di Meo G, Fiorillo C, Gianfaldoni C, Ginesini M, Hidalgo Salinas C, Lai Q, Miccoli M, Montorsi R, Pagnanelli M, Poli A, Ricci C, Sucameli F, Tamburrino D, Viti V, Cameron J, Clavien PA, Asbun HJ. REDISCOVER guidelines for borderline-resectable and locally advanced pancreatic cancer: management algorithm, unanswered questions, and future perspectives. Updates Surg 2024; 76:1573-1591. [PMID: 38684573 PMCID: PMC11455680 DOI: 10.1007/s13304-024-01860-0] [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: 03/08/2024] [Accepted: 04/10/2024] [Indexed: 05/02/2024]
Abstract
The REDISCOVER guidelines present 34 recommendations for the selection and perioperative care of borderline-resectable (BR-PDAC) and locally advanced ductal adenocarcinoma of the pancreas (LA-PDAC). These guidelines represent a significant shift from previous approaches, prioritizing tumor biology over anatomical features as the primary indication for resection. Condensed herein, they provide a practical management algorithm for clinical practice. However, the guidelines also highlight the need to redefine LA-PDAC to align with modern treatment strategies and to solve some contradictions within the current definition, such as grouping "difficult" and "impossible" to resect tumors together. Furthermore, the REDISCOVER guidelines highlight several areas requiring urgent research. These include the resection of the superior mesenteric artery, the management strategies for patients with LA-PDAC who are fit for surgery but unable to receive multi-agent neoadjuvant chemotherapy, the approach to patients with LA-PDAC who are fit for surgery but demonstrate high serum Ca 19.9 levels even after neoadjuvant treatment, and the optimal timing and number of chemotherapy cycles prior to surgery. Additionally, the role of primary chemoradiotherapy versus chemotherapy alone in LA-PDAC, the timing of surgical resection post-neoadjuvant/primary chemoradiotherapy, the efficacy of ablation therapies, and the management of oligometastasis in patients with LA-PDAC warrant investigation. Given the limited evidence for many issues, refining existing management strategies is imperative. The establishment of the REDISCOVER registry ( https://rediscover.unipi.it/ ) offers promise of a unified research platform to advance understanding and improve the management of BR-PDAC and LA-PDAC.
Collapse
Affiliation(s)
- Ugo Boggi
- Division of General and Transplant Surgery, University of Pisa, Via Savi 10, 56126, Pisa, PI, Italy.
| | - Emanuele F Kauffmann
- Division of General and Transplant Surgery, University of Pisa, Via Savi 10, 56126, Pisa, PI, Italy
| | - Niccolò Napoli
- Division of General and Transplant Surgery, University of Pisa, Via Savi 10, 56126, Pisa, PI, Italy
| | - S George Barreto
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
- Division of Surgery and Perioperative Medicine, Flinders Medical Center, Beadfor Park, Australia
| | - Marc G Besselink
- Department of Surgery, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | - Thilo Hackert
- Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Mohammad Abu Hilal
- Department of Surgery, Poliambulanza Foundation Hospital, Brescia, Italy
| | - Giovanni Marchegiani
- Hepatopancreatobiliary and Liver Transplant Surgery, Department of Surgery, Oncology and Gastroenterology, DiSCOG, University of Padua, Padua, Italy
| | - Roberto Salvia
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Shailesh V Shrikhande
- Tata Memorial Centre, Gastrointestinal and HPB Service, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Mark Truty
- Department of Surgery, Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic Rochester, Rochester, MN, USA
| | - Jens Werner
- Department of General, Visceral, and Transplant Surgery, LMU, University of Munich, Munich, Germany
| | - Christopher Wolfgang
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY, USA
| | - Elisa Bannone
- Department of Surgery, Poliambulanza Foundation Hospital, Brescia, Italy
| | | | - Alice Cattelani
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | | | - Alessandro Cucchetti
- Department of Medical and Surgical Sciences - DIMEC, Alma Mater Studiorum Università di Bologna, Bologna, Italy
| | - Davide De Sio
- Gemelli Pancreatic Center, CRMPG (Advanced Pancreatic Research Center), Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Armando Di Dato
- Division of General and Transplant Surgery, University of Pisa, Via Savi 10, 56126, Pisa, PI, Italy
| | - Giovanna Di Meo
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari, Bari, Italy
| | - Claudio Fiorillo
- Gemelli Pancreatic Center, CRMPG (Advanced Pancreatic Research Center), Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Cesare Gianfaldoni
- Division of General and Transplant Surgery, University of Pisa, Via Savi 10, 56126, Pisa, PI, Italy
| | - Michael Ginesini
- Division of General and Transplant Surgery, University of Pisa, Via Savi 10, 56126, Pisa, PI, Italy
| | | | - Quirino Lai
- Department of General and Specialty Surgery, Sapienza University of Rome, AOU Policlinico Umberto I of Rome, Rome, Italy
| | - Mario Miccoli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberto Montorsi
- Department of Surgery, Amsterdam UMC, Location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | - Andrea Poli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Claudio Ricci
- Division of Pancreatic Surgery, Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, University of Bologna, IRCCS, Azienda Ospedaliero-Universitaria di Bologna (IRCCS AOUBO), Bologna, Italy
| | - Francesco Sucameli
- Department of Surgery, Poliambulanza Foundation Hospital, Brescia, Italy
| | - Domenico Tamburrino
- Division of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Milan, Italy
| | - Virginia Viti
- Division of General and Transplant Surgery, University of Pisa, Via Savi 10, 56126, Pisa, PI, Italy
| | - John Cameron
- Department of Surgery, John Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pierre-Alain Clavien
- Department of Surgery and Transplantation, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Horacio J Asbun
- Division of Hepatobiliary and Pancreas Surgery, Miami Cancer Institute, Miami, FL, USA
| |
Collapse
|
10
|
Vitorakis N, Gargalionis AN, Papavassiliou KA, Adamopoulos C, Papavassiliou AG. Precision Targeting Strategies in Pancreatic Cancer: The Role of Tumor Microenvironment. Cancers (Basel) 2024; 16:2876. [PMID: 39199647 PMCID: PMC11352254 DOI: 10.3390/cancers16162876] [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: 07/21/2024] [Revised: 08/14/2024] [Accepted: 08/17/2024] [Indexed: 09/01/2024] Open
Abstract
Pancreatic cancer demonstrates an ever-increasing incidence over the last years and represents one of the top causes of cancer-associated mortality. Cells of the tumor microenvironment (TME) interact with cancer cells in pancreatic ductal adenocarcinoma (PDAC) tumors to preserve cancer cells' metabolism, inhibit drug delivery, enhance immune suppression mechanisms and finally develop resistance to chemotherapy and immunotherapy. New strategies target TME genetic alterations and specific pathways in cell populations of the TME. Complex molecular interactions develop between PDAC cells and TME cell populations including cancer-associated fibroblasts, myeloid-derived suppressor cells, pancreatic stellate cells, tumor-associated macrophages, tumor-associated neutrophils, and regulatory T cells. In the present review, we aim to fully explore the molecular landscape of the pancreatic cancer TME cell populations and discuss current TME targeting strategies to provide thoughts for further research and preclinical testing.
Collapse
Affiliation(s)
- Nikolaos Vitorakis
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Antonios N Gargalionis
- Department of Clinical Biochemistry, 'Attikon' University General Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Kostas A Papavassiliou
- First University Department of Respiratory Medicine, 'Sotiria' Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christos Adamopoulos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
11
|
Chervu N, Kim S, Sakowitz S, Le N, Mallick S, Lee H, Benharash P, Donahue T. Disparities in neoadjuvant chemotherapy for pancreatic adenocarcinoma with vascular involvement. Surg Open Sci 2024; 20:101-105. [PMID: 39021616 PMCID: PMC11252929 DOI: 10.1016/j.sopen.2024.06.003] [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: 05/31/2024] [Accepted: 06/12/2024] [Indexed: 07/20/2024] Open
Abstract
Background Multiagent neoadjuvant chemotherapy (NAT) has been linked with improved survival for locally advanced (LA) or borderline resectable (BR) pancreatic ductal adenocarcinoma (PDAC). However, the existence of disparities in its utilization remains to be elucidated. Methods All adults with PDAC were tabulated from the 2011-2017 Nationwide Cancer Database. Tumor vascular involvement was determined using the clinical T stage and CS_EXTENSION variables. The significance of temporal trends was calculated using Cuzick's non-parametric test. A Cox proportional hazard model was used to assess the impact of NAT utilization on hazard of two-year mortality. A logistic regression model was developed to determine factors associated with receipt of NAT. Results Of 3811 patients meeting inclusion criteria, 50.8 % received NAT. NAT utilization significantly increased over the study period, from 31.7 % in 2011 to 81.1 % in 2017 (p < 0.001). NAT was associated with significantly reduced two-year mortality (Hazards Ratio 0.34, 95 % Confidence Interval [CI] 0.18-0.67).After adjustment, younger (Adjusted Odds Ratio [AOR] 0.97/year, CI 0.96-0.98) and Black (AOR 0.65, CI 0.48-0.89; ref: White) patients demonstrated reduced odds of NAT. Furthermore, patients with Medicare (AOR 0.73, CI 0.59-0.90; ref: Private) or Medicaid insurance (AOR 0.67, CI 0.46-0.97; ref: Private) had lower odds of NAT, as did those treated at non-academic institutions (Community: AOR 0.42, CI 0.35-0.52, Integrated: 0.68, CI 0.54-0.85) or in the lowest education quartile (AOR 0.52, CI 0.29-0.95; ref: Highest). Conclusions We identified increasing utilization of NAT for BR/LA pancreatic adenocarcinoma. Despite being linked with significantly reduced two-year mortality, socioeconomic disparities affect odds of NAT.
Collapse
Affiliation(s)
- Nikhil Chervu
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Shineui Kim
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Sara Sakowitz
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Nguyen Le
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Saad Mallick
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Hanjoo Lee
- Department of Surgery, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Peyman Benharash
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Timothy Donahue
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, UCLA, Los Angeles, CA, USA
| |
Collapse
|
12
|
Fang Z, Xu X, Tao L, Lino-Silva LS, Lu Y. MMP11 as a prognostic biomarker correlated with immune infiltrates in pancreatic adenocarcinoma. J Gastrointest Oncol 2024; 15:1224-1244. [PMID: 38989433 PMCID: PMC11231847 DOI: 10.21037/jgo-24-425] [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: 06/04/2024] [Accepted: 06/24/2024] [Indexed: 07/12/2024] Open
Abstract
Background Matrix metalloproteinase 11 (MMP11) plays a vital role in cell proliferation, apoptosis, tumor angiogenesis, migration, and other basic processes. Currently, few studies have examined the value of MMP11 in pancreatic cancer in relation to prognostic risk, diagnostic indicators, and immunotherapy. This study aims to explore the association between MMP11 and the tumor immune microenvironment in pancreatic adenocarcinoma (PAAD). Methods We selected clinical samples and data downloaded from The Cancer Genome Atlas and Genotype-Tissue Expression, in addition, we use other online data for further analysis. Through a comprehensive bioinformatics investigation, we systematically analyzed the clinical significance and expression level of MMP11 in pancreatic cancer. Results MMP11 was overexpressed in many cancers, and a higher expression of MMP11 was associated with a poorer prognosis in pancreatic cancer. Conversely, the hypermethylation of MMP11 was associated with better overall survival. The MMP11 expression network had widespread effects on the prognosis and immune activation of PAAD. The expression of MMP11 was significantly associated with a variety of tumor-infiltrating immune cells. An association was also found between MMP11 expression and chemokines in PAAD. High MMP11 expression might be involved in immune cell migration to the tumor microenvironment. Conclusions MMP11 is a prognostic biomarker for patients in pancreatic cancer and may regulate the tumor immune microenvironment. The potential effects and mechanisms of MMP11 in PAAD require further exploring.
Collapse
Affiliation(s)
- Zhengxuying Fang
- Department of Health Science Center, Medical College of Ningbo University, Ningbo, China
| | - Xiaoyu Xu
- Department of Oncology, The Yuyao People's Hospital, Ningbo, China
| | - Linglong Tao
- Department of Health Science Center, Medical College of Ningbo University, Ningbo, China
| | - Leonardo S Lino-Silva
- Gastrointestinal Pathology, Instituto Nacional de Cancerología de México (Mexico's National Cancer Institute), Mexico City, Mexico
| | - Yi Lu
- Department of Radiation Oncology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
13
|
Zhu C, Wang C, Wang X, Dong S, Xu Q, Zheng J. PABPC1 silencing inhibits pancreatic cancer cell proliferation and EMT, and induces apoptosis via PI3K/AKT pathway. Cytotechnology 2024; 76:351-361. [PMID: 38736728 PMCID: PMC11082123 DOI: 10.1007/s10616-024-00626-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 03/11/2024] [Indexed: 05/14/2024] Open
Abstract
Pancreatic cancer is difficult to manage owing to the challenges involved in its treatment and nursing. This study aimed to clarify the roles and mechanisms of action of Poly (A)-binding protein cytoplasmic 1 (PABPC1) on pancreatic cancer. The expression of PABPC1 in pancreatic cancer tissues and cell lines was detected using RT-qPCR and western blotting. The effects of PABPC1 on proliferation, apoptosis, epithelial-mesenchymal transition (EMT), and the PI3K/AKT signaling pathway in pancreatic cancer cells were further investigated using MTT assays, flow cytometry, and western blotting. The expression of PABPC1 was significantly upregulated in pancreatic cancer tissues and cells, whereas PABPC1 downregulation inhibited pancreatic cancer cell proliferation, induced apoptosis, decreased the expression of EMT-associated proteins, and exerted a regulatory effect by inhibiting the PI3K/AKT signaling pathway. In addition, the findings indicated that PABPC1 over-expression significantly promoted pancreatic cancer cell proliferation, inhibited apoptosis, decreased the expression of E-cadherin, enhanced N-cadherin expression, and activating the PI3K/AKT signaling pathway. PABPC1 silencing significantly inhibited proliferation and EMT and induced apoptosis in pancreatic cancer cells. These findings provide novel insights into the role of PABPC1 in the development of pancreatic cancer. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-024-00626-1.
Collapse
Affiliation(s)
- Changren Zhu
- Pathology Department, Northern Jiangsu People’s Hospital of Jiangsu Province, Yangzhou, 225001 China
| | - Cuimei Wang
- Pathology Department, Northern Jiangsu People’s Hospital of Jiangsu Province, Yangzhou, 225001 China
| | - Xiaodong Wang
- Department of Biliary and Pancreatic Surgery, Northern Jiangsu People’s Hospital of Jiangsu Province, Yangzhou, 225001 China
| | - Shuangshuang Dong
- Pathology Department, Northern Jiangsu People’s Hospital of Jiangsu Province, Yangzhou, 225001 China
| | - Qing Xu
- Pathology Department, Northern Jiangsu People’s Hospital of Jiangsu Province, Yangzhou, 225001 China
| | - Jun Zheng
- Daytime Surgical Ward, Northern Jiangsu People’s Hospital of Jiangsu Province, No. 98 Nantong West Road, Yangzhou, 225001 China
| |
Collapse
|
14
|
Kandathil A, Subramaniam R. Quarter-Century PET/Computed Tomography Transformation of Oncology: Hepatobiliary and Pancreatic Cancer. PET Clin 2024; 19:163-175. [PMID: 38212214 DOI: 10.1016/j.cpet.2023.12.003] [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: 01/13/2024]
Abstract
[18F] Fluorodeoxyglucose (18F-FDG) PET/CT can improve the staging accuracy and clinical management of patients with hepatobiliary and pancreatic cancers, by detection of unsuspected metastases. 18F-FDG PET/CT metabolic parameters are valuable in predicting treatment response and survival. Metabolic response on 18F-FDG PET/CT can predict preoperative pathologic response to neoadjuvant therapy in patients with pancreatic cancer and determine prognosis. Several novel non-FDG tracers, such as 68Ga prostate-specific membrane antigen (PSMA) and 68Ga-fibroblast activation protein inhibitor (FAPI) PET/CT, show promise for imaging hepatobiliary and pancreatic cancers with potential for radioligand therapy.
Collapse
Affiliation(s)
- Asha Kandathil
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Rathan Subramaniam
- Faculty of Medicine, Nursing, Midwifery and Health Sciences, University of Notre Dame Australia, Sydney, Australia; Department of Radiology, Duke University, Durham, NC, USA; Department of Medicine, University of Otago Medical School, Dunedin, New Zealand
| |
Collapse
|
15
|
Song N, Cui K, Zeng L, Li M, Fan Y, Shi P, Wang Z, Su W, Wang H. Advance in the role of chemokines/chemokine receptors in carcinogenesis: Focus on pancreatic cancer. Eur J Pharmacol 2024; 967:176357. [PMID: 38309677 DOI: 10.1016/j.ejphar.2024.176357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/05/2024]
Abstract
The chemokines/chemokine receptors pathway significantly influences cell migration, particularly in recruiting immune cells to the tumor microenvironment (TME), impacting tumor progression and treatment outcomes. Emerging research emphasizes the involvement of chemokines in drug resistance across various tumor therapies, including immunotherapy, chemotherapy, and targeted therapy. This review focuses on the role of chemokines/chemokine receptors in pancreatic cancer (PC) development, highlighting their impact on TME remodeling, immunotherapy, and relevant signaling pathways. The unique immunosuppressive microenvironment formed by the interaction of tumor cells, stromal cells and immune cells plays an important role in the tumor proliferation, invasion, migration and therapeutic resistance. Chemokines/chemokine receptors, such as chemokine ligand (CCL) 2, CCL3, CCL5, CCL20, CCL21, C-X-C motif chemokine ligand (CXCL) 1, CXCL2, CXCL3, CXCL4, CXCL5, CXCL8, CXCL9, CXCL10, CXCL11, CXCL12, CXCL13, CXCL14, CXCL16, CXCL17, and C-X3-C motif chemokine ligand (CX3CL)1, derived mainly from leukocyte cells, cancer-related fibroblasts (CAFs), pancreatic stellate cells (PSCs), and tumor-associated macrophages (TAMs), contribute to PC progression and treatment resistance. Chemokines recruit myeloid-derived suppressor cells (MDSC), regulatory T cells (Tregs), and M2 macrophages, inhibiting the anti-tumor activity of immune cells. Simultaneously, they enhance pathways like epithelial-mesenchymal transition (EMT), Akt serine/threonine kinase (AKT), extracellular regulated protein kinases (ERK) 1/2, and nuclear factor kappa-B (NF-κB), etc., elevating the risk of PC metastasis and compromising the efficacy of radiotherapy, chemotherapy, and anti-PD-1/PD-L1 immunotherapy. Notably, the CCLx-CCR2 and CXCLx-CXCR2/4 axis emerge as potential therapeutic targets in PC. This review integrates recent findings on chemokines and receptors in PC treatment, offering valuable insights for innovative therapeutic approaches.
Collapse
Affiliation(s)
- Na Song
- Department of Pathology, Xinxiang Key Laboratory of Precision Medicine, The First Affiliated Hospital of Xinxiang Medical University, China; Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Kai Cui
- Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Liqun Zeng
- Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Mengxiao Li
- Department of Pathology, Xinxiang Key Laboratory of Precision Medicine, The First Affiliated Hospital of Xinxiang Medical University, China
| | - Yanwu Fan
- Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Pingyu Shi
- Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Ziwei Wang
- Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China
| | - Wei Su
- Department of Pathology, Xinxiang Key Laboratory of Precision Medicine, The First Affiliated Hospital of Xinxiang Medical University, China.
| | - Haijun Wang
- Department of Pathology, Xinxiang Key Laboratory of Precision Medicine, The First Affiliated Hospital of Xinxiang Medical University, China; Department of Pathology, Xinxiang Medical University, Xinxiang, 453000, China.
| |
Collapse
|
16
|
He Y, Zhao Y, Akhtar ML, Li Y, E M, Nie H. Neoadjuvant therapy for non-small cell lung cancer and esophageal cancer. Am J Cancer Res 2024; 14:1258-1277. [PMID: 38590425 PMCID: PMC10998743 DOI: 10.62347/tcec1867] [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: 11/14/2023] [Accepted: 03/13/2024] [Indexed: 04/10/2024] Open
Abstract
As the major malignant tumors in the chest, non-small cell lung cancer (NSCLC) and esophageal cancer (EC) bring huge health burden to human beings worldwide. Currently, surgery is still the mainstay for comprehensive treatment for NSCLC and EC, but the prognosis is still poor as the results of cancer recurrence and distant metastasis. Neoadjuvant therapy refers to a single or combined treatment before surgery, aiming to improve the therapeutic effects of the traditional therapies. Unfortunately, the clinical outcomes and effects of neoadjuvant therapy are still controversial due to its apparent advantages and disadvantages, and different patients may respond differentially to the same scheme of neoadjuvant therapy, which makes it urgent and necessary to develop personalized scheme of neoadjuvant therapy for different individuals. Therefore, this review summarizes the novel schemes and strategies of neoadjuvant therapy, which may help to significantly improve of life quality of patients suffering from chest-related malignancies.
Collapse
Affiliation(s)
- Yunlong He
- School of Life Science and Technology, Harbin Institute of TechnologyHarbin 150008, Heilongjiang, China
- Department of Radiation Oncology, Harbin Medical University Cancer HospitalHarbin 150060, Heilongjiang, China
| | - Yaqi Zhao
- School of Life Science and Technology, Harbin Institute of TechnologyHarbin 150008, Heilongjiang, China
| | - Muhammad Luqman Akhtar
- School of Life Science and Technology, Harbin Institute of TechnologyHarbin 150008, Heilongjiang, China
| | - Yu Li
- School of Life Science and Technology, Harbin Institute of TechnologyHarbin 150008, Heilongjiang, China
| | - Mingyan E
- Department of Radiation Oncology, Harbin Medical University Cancer HospitalHarbin 150060, Heilongjiang, China
| | - Huan Nie
- School of Life Science and Technology, Harbin Institute of TechnologyHarbin 150008, Heilongjiang, China
| |
Collapse
|
17
|
Čebron Ž, Djokić M, Petrič M, Čemažar M, Bošnjak M, Serša G, Trotovšek B. Intraoperative electrochemotherapy of the posterior resection surface after pancreaticoduodenectomy: Preliminary results of a hybrid approach treatment of pancreatic cancer. Bioelectrochemistry 2024; 155:108576. [PMID: 37748261 DOI: 10.1016/j.bioelechem.2023.108576] [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: 06/11/2023] [Revised: 09/15/2023] [Accepted: 09/17/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND Despite extensive research in recent decades, pancreatic cancer continues to be among the most lethal forms of cancer, with no substantial increase in survival rates. Local recurrences account for approximately 30 per cent of all disease recurrences. With the intent to improve survival, we designed a novel, hybrid treatment strategy consisting of surgical resection and additional intraoperative electrochemotherapy of the posterior resection surface. We present the study protocols and preliminary findings of a prospective pilot study investigating this treatment approach. METHODS Consenting patients with resectable pancreatic head ductal adenocarcinoma who met the inclusion criteria were enrolled in the study. After surgical resection, electrochemotherapy with bleomycin was performed using plate electrodes to cover the area between anatomical landmarks. RESULTS Electrochemotherapy of the posterior resection surface was feasible in all 7 patients. We observed pancreatic fistula grade B in only one patient; all other noted complications were Clavien-Dindo grade 2 or less. The hospital mortality was 0%. CONCLUSIONS Our preliminary results suggest that a hybrid approach combining surgery with intraoperative electrochemotherapy is safe and feasible.
Collapse
Affiliation(s)
- Žan Čebron
- University Medical Centre Ljubljana, Department of Abdominal Surgery, Zaloska 7, SI-1000 Ljubljana, Slovenia; University of Ljubljana, Medical Faculty, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Mihajlo Djokić
- University Medical Centre Ljubljana, Department of Abdominal Surgery, Zaloska 7, SI-1000 Ljubljana, Slovenia; University of Ljubljana, Medical Faculty, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Miha Petrič
- University Medical Centre Ljubljana, Department of Abdominal Surgery, Zaloska 7, SI-1000 Ljubljana, Slovenia; University of Ljubljana, Medical Faculty, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Maja Čemažar
- Institute of Oncology Ljubljana, Department of Experimental Oncology, Zaloska 2, SI-1000 Ljubljana, Slovenia; University of Primorska, Faculty of Health Sciences, Polje 42, SI-6310 Izola, Slovenia
| | - Maša Bošnjak
- Institute of Oncology Ljubljana, Department of Experimental Oncology, Zaloska 2, SI-1000 Ljubljana, Slovenia
| | - Gregor Serša
- Institute of Oncology Ljubljana, Department of Experimental Oncology, Zaloska 2, SI-1000 Ljubljana, Slovenia; University of Ljubljana, Faculty of Health Sciences, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenia.
| | - Blaž Trotovšek
- University Medical Centre Ljubljana, Department of Abdominal Surgery, Zaloska 7, SI-1000 Ljubljana, Slovenia; University of Ljubljana, Medical Faculty, Vrazov trg 2, 1000 Ljubljana, Slovenia.
| |
Collapse
|
18
|
Zhao B, Xia C, Xia T, Qiu Y, Zhu L, Cao B, Gao Y, Ge R, Cai W, Ding Z, Yu Q, Lu C, Tang T, Wang Y, Song Y, Long X, Ye J, Lu D, Ju S. Development of a radiomics-based model to predict occult liver metastases of pancreatic ductal adenocarcinoma: a multicenter study. Int J Surg 2024; 110:740-749. [PMID: 38085810 PMCID: PMC10871636 DOI: 10.1097/js9.0000000000000908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 11/02/2023] [Indexed: 02/17/2024]
Abstract
BACKGROUND Undetectable occult liver metastases block the long-term survival of pancreatic ductal adenocarcinoma (PDAC). This study aimed to develop a radiomics-based model to predict occult liver metastases and assess its prognostic capacity for survival. MATERIALS AND METHODS Patients who underwent surgical resection and were pathologically proven with PDAC were recruited retrospectively from five tertiary hospitals between January 2015 and December 2020. Radiomics features were extracted from tumors, and the radiomics-based model was developed in the training cohort using LASSO-logistic regression. The model's performance was assessed in the internal and external validation cohorts using the area under the receiver operating curve (AUC). Subsequently, the association of the model's risk stratification with progression-free survival (PFS) and overall survival (OS) was then statistically examined using Cox regression analysis and the log-rank test. RESULTS A total of 438 patients [mean (SD) age, 62.0 (10.0) years; 255 (58.2%) male] were divided into the training cohort ( n =235), internal validation cohort ( n =100), and external validation cohort ( n =103). The radiomics-based model yielded an AUC of 0.73 (95% CI: 0.66-0.80), 0.72 (95% CI: 0.62-0.80), and 0.71 (95% CI: 0.61-0.80) in the training, internal validation, and external validation cohorts, respectively, which were higher than the preoperative clinical model. The model's risk stratification was an independent predictor of PFS (all P <0.05) and OS (all P <0.05). Furthermore, patients in the high-risk group stratified by the model consistently had a significantly shorter PFS and OS at each TNM stage (all P <0.05). CONCLUSION The proposed radiomics-based model provided a promising tool to predict occult liver metastases and had a great significance in prognosis.
Collapse
Affiliation(s)
- Ben Zhao
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Cong Xia
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Tianyi Xia
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Yue Qiu
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Liwen Zhu
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Buyue Cao
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Yin Gao
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Rongjun Ge
- School of Instrument Science and Engineering, Southeast University, Nanjing
| | - Wu Cai
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou
| | - Zhimin Ding
- Department of Radiology, Yijishan Hospital of Wannan Medical College, Wuhu
| | - Qian Yu
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Chunqiang Lu
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Tianyu Tang
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Yuancheng Wang
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| | - Yang Song
- MR Scientific Marketing, Siemens Healthineers, Shanghai
| | - Xueying Long
- Department of Radiology, The Xiangya Hospital of Central South University, Changsha
| | - Jing Ye
- Department of Radiology, Northern Jiangsu People’s Hospital, Yangzhou
| | - Dong Lu
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, People’s Republic of China
| | - Shenghong Ju
- Department of Radiology, The Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, School of Medicine
| |
Collapse
|
19
|
Wilbur HC, Soares HP, Azad NS. Neoadjuvant and adjuvant therapy for biliary tract cancer: Advances and limitations. Hepatology 2024:01515467-990000000-00725. [PMID: 38266282 DOI: 10.1097/hep.0000000000000760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/31/2023] [Indexed: 01/26/2024]
Abstract
Biliary tract cancers (BTC) are a rare and aggressive consortium of malignancies, consisting of intrahepatic cholangiocarcinoma, extrahepatic cholangiocarcinoma, and gallbladder carcinoma. While most patients present with metastatic disease, a minority of patients with BTC are eligible for curative surgical resection at the time of presentation. However, these patients have poor 5-year overall survival rates and high rates of recurrence, necessitating the improvement of the neoadjuvant and adjuvant treatment of BTC. In this review, we assess the neoadjuvant and adjuvant clinical trials for the treatment of BTC and discuss the challenges and limitations of clinical trials, as well as future directions for the treatment of BTC.
Collapse
Affiliation(s)
- H Catherine Wilbur
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Heloisa P Soares
- Division of Oncology, Department of Internal Medicine Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Nilofer S Azad
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
20
|
Luo X, Ye Z, Xu C, Chen H, Dai S, Chen W, Bao G. Corosolic acid enhances oxidative stress-induced apoptosis and senescence in pancreatic cancer cells by inhibiting the JAK2/STAT3 pathway. Mol Biol Rep 2024; 51:176. [PMID: 38252208 DOI: 10.1007/s11033-023-09105-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/05/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Pancreatic cancer (PC) is a fatal human malignancy with a poor prognosis. Corosolic acid (CRA) is a triterpenoid, has been reported to have inhibitory effects on tumor growth. However, the role of CRA on PC has not been explored. Here, we aimed to uncover the molecular mechanisms of CRA in PC progression. METHODS Cell viability, lactate dehydrogenase (LDH) release, cell apoptosis and senescence were detected by cell counting kit-8 (CCK-8), LDH, flow cytometry and senescence associated-β-galactosidase (SA-β-gal) assay. Levels of relevant proteins and oxidative stress (OS) markers were evaluated by Western blot and enzyme-linked immunosorbent assay (ELISA). A xenograft tumor model was established to explore the in vivo effects of CRA on PC. RESULTS We found that CRA inhibited PC cell viability and promoted LDH release in a dose-dependent manner, but had no significant effect on human normal pancreatic ductal epithelial cells HPDE6C7. CRA increased OS-induced cell apoptosis and senescence in HAPC and SW1990 cells. And CRA decreased the levels of anti-apoptotic protein Bcl-2, and elevated the expression of pro-apoptotic protein Bax and senescence-associated proteins P21 and P53. Besides, CRA decreased tumor growth in xenograft models. Furthermore, CRA inactivated the Janus kinase-2 (JAK2)/Signal Transducer and Activator of Transcription 3 (STAT3) signaling pathway in HAPC and SW1990 cells. Functional experiments demonstrated that activation of the JAK2/STAT3 pathway by the JAK2 activator coumermycin A1 (C-A1) or the STAT3 activator colivelin (col) reduced the contribution effect of OS, apoptosis and senescence by CRA. CONCLUSION Taken together, our findings indicated that CRA exerted anti-cancer effects in PC by inhibiting the JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Xu Luo
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Kunming, 650032, Yunnan Province, China
| | - Zhengchen Ye
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Kunming, 650032, Yunnan Province, China
| | - Chenglei Xu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Kunming, 650032, Yunnan Province, China
| | - Huan Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Kunming, 650032, Yunnan Province, China
| | - Shupeng Dai
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Kunming, 650032, Yunnan Province, China
| | - Weihong Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Kunming, 650032, Yunnan Province, China
| | - Guoqing Bao
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Kunming, 650032, Yunnan Province, China.
| |
Collapse
|
21
|
Huang XD, Chen YW, Tian L, Du L, Cheng XC, Lu YX, Lin DD, Xiao FJ. NUDT21 interacts with NDUFS2 to activate the PI3K/AKT pathway and promotes pancreatic cancer pathogenesis. J Cancer Res Clin Oncol 2024; 150:8. [PMID: 38195952 PMCID: PMC10776698 DOI: 10.1007/s00432-023-05540-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 11/29/2023] [Indexed: 01/11/2024]
Abstract
BACKGROUND NUDT21 (Nudix Hydrolase 21) has been shown to play an essential role in multiple biological processes. Pancreatic adenocarcinoma (PAAD) is one of the most fatal cancers in the world. However, the biological function of NUDT21 in PAAD remains rarely understood. The aim of this research was to identify the prediction value of NUDT21 in diagnosis, prognosis, immune infiltration, and signal pathway in PAAD. METHODS Combined with the data in online databases, we analyzed the expression, immune infiltration, function enrichment, signal pathway, diagnosis, and prognosis of NUDT21 in PAAD. Then, the biological function of NUDT21 and its interacted protein in PAAD was identified through plasmid transduction system and protein mass spectrometry. Expression of NUDT21 was further verified in clinical specimens by immunofluorescence. RESULTS We found that NUDT21 was upregulated in PAAD tissues and was significantly associated with the diagnosis and prognosis of pancreatic cancer through bioinformatic data analysis. We also found that overexpression of NUDT21 enhanced PAAD cells proliferation and migration, whereas knockdown NUDT21 restored the effects through in vitro experiment. Moreover, NDUFS2 was recognized as a potential target of NUDT21.We further verified that the expression of NDUFS2 was positively correlated with NUDT21 in PAAD clinical specimens. Mechanically, we found that NUDT21 stabilizes NDUFS2 and activates the PI3K-AKT signaling pathway. CONCLUSION Our investigation reveals that NUDT21 is a previously unrecognized oncogenic factor in the diagnosis, prognosis, and treatment target of PAAD, and we suggest that NUDT21 might be a novel therapeutic target in PAAD.
Collapse
Affiliation(s)
- Xiao-Dong Huang
- Department of General Surgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, People's Republic of China
| | - Yong-Wei Chen
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center of PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Lv Tian
- School of Nursing, Jilin University, Changchun, 130015, People's Republic of China
| | - Li Du
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Xiao-Chen Cheng
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Yu-Xin Lu
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Dong-Dong Lin
- Department of General Surgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, People's Republic of China.
| | - Feng-Jun Xiao
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.
| |
Collapse
|
22
|
Pacella G, Brunese MC, D’Imperio E, Rotondo M, Scacchi A, Carbone M, Guerra G. Pancreatic Ductal Adenocarcinoma: Update of CT-Based Radiomics Applications in the Pre-Surgical Prediction of the Risk of Post-Operative Fistula, Resectability Status and Prognosis. J Clin Med 2023; 12:7380. [PMID: 38068432 PMCID: PMC10707069 DOI: 10.3390/jcm12237380] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 09/10/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is the seventh leading cause of cancer-related deaths worldwide. Surgical resection is the main driver to improving survival in resectable tumors, while neoadjuvant treatment based on chemotherapy (and radiotherapy) is the best option-treatment for a non-primally resectable disease. CT-based imaging has a central role in detecting, staging, and managing PDAC. As several authors have proposed radiomics for risk stratification in patients undergoing surgery for PADC, in this narrative review, we have explored the actual fields of interest of radiomics tools in PDAC built on pre-surgical imaging and clinical variables, to obtain more objective and reliable predictors. METHODS The PubMed database was searched for papers published in the English language no earlier than January 2018. RESULTS We found 301 studies, and 11 satisfied our research criteria. Of those included, four were on resectability status prediction, three on preoperative pancreatic fistula (POPF) prediction, and four on survival prediction. Most of the studies were retrospective. CONCLUSIONS It is possible to conclude that many performing models have been developed to get predictive information in pre-surgical evaluation. However, all the studies were retrospective, lacking further external validation in prospective and multicentric cohorts. Furthermore, the radiomics models and the expression of results should be standardized and automatized to be applicable in clinical practice.
Collapse
Affiliation(s)
- Giulia Pacella
- Department of Medicine and Health Science “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (G.P.)
| | - Maria Chiara Brunese
- Department of Medicine and Health Science “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (G.P.)
| | | | - Marco Rotondo
- Department of Medicine and Health Science “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (G.P.)
| | - Andrea Scacchi
- General Surgery Unit, University of Milano-Bicocca, 20126 Milan, Italy
| | - Mattia Carbone
- San Giovanni di Dio e Ruggi d’Aragona Hospital, 84131 Salerno, Italy;
| | - Germano Guerra
- Department of Medicine and Health Science “V. Tiberio”, University of Molise, 86100 Campobasso, Italy; (G.P.)
| |
Collapse
|
23
|
Sun LP, Bai WQ, Zhou DD, Wu XF, Zhang LW, Cui AL, Xie ZH, Gao RJ, Zhen YS, Li ZR, Miao QF. hIMB1636-MMAE, a Novel TROP2-Targeting Antibody-Drug Conjugate Exerting Potent Antitumor Efficacy in Pancreatic Cancer. J Med Chem 2023; 66:14700-14715. [PMID: 37883180 DOI: 10.1021/acs.jmedchem.3c01210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
Herein, we first prepared a novel anti-TROP2 antibody-drug conjugate (ADC) hIMB1636-MMAE using hIMB1636 antibody chemically coupled to monomethyl auristatin E (MMAE) via a Valine-Citrulline linker and then reported its characteristics and antitumor activity. With a DAR of 3.92, it binds specifically to both recombinant antigen (KD ∼ 0.687 nM) and cancer cells and could be internalized by target cells and selectively kill them with IC50 values at nanomolar/subnanomolar levels by inducing apoptosis and G2/M phase arrest. hIMB1636-MMAE also inhibited cell migration, induced ADCC effects, and had bystander effects. It displayed significant tumor-targeting ability and excellent tumor-suppressive effects in vivo, resulting in 5/8 tumor elimination at 12 mg/kg in the T3M4 xenograft model or complete tumor disappearance at 10 mg/kg in BxPc-3 xenografts in nude mice. Its half-life in mice was about 87 h. These data suggested that hIMB1636-MMAE was a promising candidate for the treatment of pancreatic cancer with TROP2 overexpression.
Collapse
Affiliation(s)
- Li-Ping Sun
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Tiantan Xili, Beijing 100050, China
| | - Wei-Qi Bai
- Department of Organic Chemistry, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Tiantan Xili, Beijing 100050, China
| | - Dan-Dan Zhou
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Tiantan Xili, Beijing 100050, China
| | - Xiao-Fan Wu
- Department of Organic Chemistry, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Tiantan Xili, Beijing 100050, China
| | - Lan-Wen Zhang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Tiantan Xili, Beijing 100050, China
| | - A-Long Cui
- Department of Organic Chemistry, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Tiantan Xili, Beijing 100050, China
| | - Zi-Hui Xie
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Tiantan Xili, Beijing 100050, China
| | - Rui-Juan Gao
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Tiantan Xili, Beijing 100050, China
| | - Yong-Su Zhen
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Tiantan Xili, Beijing 100050, China
| | - Zhuo-Rong Li
- Department of Organic Chemistry, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Tiantan Xili, Beijing 100050, China
| | - Qing-Fang Miao
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Tiantan Xili, Beijing 100050, China
| |
Collapse
|
24
|
Zhou B, Zhang SR, Chen G, Chen P. Developments and challenges in neoadjuvant therapy for locally advanced pancreatic cancer. World J Gastroenterol 2023; 29:5094-5103. [PMID: 37744290 PMCID: PMC10514760 DOI: 10.3748/wjg.v29.i35.5094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/19/2023] [Accepted: 08/31/2023] [Indexed: 09/14/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a significant public health challenge and is currently the fourth leading cause of cancer-related mortality in developed countries. Despite advances in cancer treatment, the 5-year survival rate for patients with PDAC remains less than 5%. In recent years, neoadjuvant therapy (NAT) has emerged as a promising treatment option for many cancer types, including locally advanced PDAC, with the potential to improve patient outcomes. To analyze the role of NAT in the setting of locally advanced PDAC over the past decade, a systematic literature search was conducted using PubMed and Web of Science. The results suggest that NAT may reduce the local mass size, promote tumor downstaging, and increase the likelihood of resection. These findings are supported by the latest evidence-based medical literature and the clinical experience of our center. Despite the potential benefits of NAT, there are still challenges that need to be addressed. One such challenge is the lack of consensus on the optimal timing and duration of NAT. Improved criteria for patient selection are needed to further identify PDAC patients likely to respond to NAT. In conclusion, NAT has emerged as a promising treatment option for locally advanced PDAC. However, further research is needed to optimize its use and to better understand the role of NAT in the management of this challenging disease. With continued advances in cancer treatment, there is hope of improving the outcomes of patients with PDAC in the future.
Collapse
Affiliation(s)
- Bo Zhou
- Department of Hepatobiliary Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Shi-Ran Zhang
- Department of Hepatobiliary Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Geng Chen
- Department of Hepatobiliary Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Ping Chen
- Department of Hepatobiliary Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| |
Collapse
|
25
|
Wang F, Guo H, Li S, Xu J, Yu D. The value of enhanced CT features and texture-signatures in assessing the inflammatory infiltration of pancreatic ductal adenocarcinoma. Front Oncol 2023; 13:1078861. [PMID: 36816950 PMCID: PMC9936180 DOI: 10.3389/fonc.2023.1078861] [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: 10/24/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Purpose To explore the predictive value of computed tomography (CT) imaging features and CT-based texture analysis in assessing inflammatory infiltration in pancreatic ductal adenocarcinoma (PDAC). Methods A total of 43 patients with PDAC confirmed by surgical pathology were included in the study. The clinical, radiological, surgical, and pathological features of the patients were analyzed retrospectively using the chi-square test or Spearman's correlation. Receiver operating characteristic (ROC) curves were utilized to assess the overall predictive ability of the tumor enhancement degree on triphasic contrast-enhanced CT images for the inflammatory infiltration degree in PDAC. Furthermore, all CT data were uploaded to the RadCloud platform for region of interest (ROI) delineation and feature extraction. Then, the Variance Threshold and SelectKBest algorithms were used to find the optimal CT features. Binary logistic regression was employed to analyze the selected features in all three contrast-enhanced CT phases, and regression equations were formulated. ROC analysis was performed to evaluate the predictive effectiveness of each equation. Results The analysis revealed a statistically significant correlation between the degree of differentiation and radiological findings such as necrosis and cystic degeneration, vascular invasion, and the presence of ascites (P < 0.05). The enhancement degree of the tumor in both the arterial and venous phases was significantly correlated with the inflammatory infiltration degree (P < 0.05); however, the areas under the ROC curve (AUCs) of arterial and venous enhancement were 0.570 and 0.542, respectively. Regression equations based on the texture features of triphasic contrast-enhanced tumors were formulated, and their AUCs were 0.982, 0.643, and 0.849. Conclusion Conventional radiological features are not significantly correlated with the degree of inflammatory infiltration in PDAC. The enhancement degrees in both the arterial phase and venous phase were statistically correlated with the inflammatory infiltration level but had poor predictive value. The texture features of PDAC on contrast-enhanced CT may show a better assessment value, especially in the arterial phase.
Collapse
Affiliation(s)
- Fangqing Wang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Hang Guo
- Department of Radiology, Laiyang Central Hospital of Yantai, Yantai, China
| | - Shunjia Li
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Jianwei Xu
- Department of Pancreatic Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Dexin Yu
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China,*Correspondence: Dexin Yu,
| |
Collapse
|
26
|
Yang P, Mao K, Gao Y, Wang Z, Wang J, Chen Y, Ma C, Bian Y, Shao C, Lu J. Tumor size measurements of pancreatic cancer with neoadjuvant therapy based on RECIST guidelines: is MRI as effective as CT? Cancer Imaging 2023; 23:8. [PMID: 36653861 PMCID: PMC9850516 DOI: 10.1186/s40644-023-00528-z] [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: 10/10/2022] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVES To compare tumor size measurements using CT and MRI in pancreatic cancer (PC) patients with neoadjuvant therapy (NAT). METHODS This study included 125 histologically confirmed PC patients who underwent NAT. The tumor sizes from CT and MRI before and after NAT were compared by using Bland-Altman analyses and intraclass correlation coefficients (ICCs). Variations in tumor size estimates between MRI and CT in relationship to different factors, including NAT methods (chemotherapy, chemoradiotherapy), tumor locations (head/neck, body/tail), tumor regression grade (TRG) levels (0-2, 3), N stages (N0, N1/N2) and tumor resection margin status (R0, R1), were further analysed. The McNemar test was used to compare the efficacy of NAT evaluations based on the CT and MRI measurements according to RECIST 1.1 criteria. RESULTS There was no significant difference between the median tumor sizes from CT and MRI before and after NAT (P = 0.44 and 0.39, respectively). There was excellent agreement in tumor size between MRI and CT, with mean size differences and limits of agreement (LOAs) of 1.5 [-9.6 to 12.7] mm and 0.9 [-12.6 to 14.5] mm before NAT (ICC, 0.93) and after NAT (ICC, 0.91), respectively. For all the investigated factors, there was good or excellent correlation (ICC, 0.76 to 0.95) for tumor sizes between CT and MRI. There was no significant difference in the efficacy evaluation of NAT between CT and MRI measurements (P = 1.0). CONCLUSION MRI and CT have similar performance in assessing PC tumor size before and after NAT.
Collapse
Affiliation(s)
- Panpan Yang
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China
| | - Kuanzheng Mao
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China ,grid.267139.80000 0000 9188 055XSchool of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yisha Gao
- grid.73113.370000 0004 0369 1660Department of Pathology, Changhai Hospital of Shanghai, Naval Medical University, Shanghai, China
| | - Zhen Wang
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China
| | - Jun Wang
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China
| | - Yufei Chen
- grid.24516.340000000123704535College of Electronic and Information Engineering, Tongji University, Shanghai, China
| | - Chao Ma
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China ,grid.24516.340000000123704535College of Electronic and Information Engineering, Tongji University, Shanghai, China
| | - Yun Bian
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China
| | - Chengwei Shao
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China
| | - Jianping Lu
- grid.73113.370000 0004 0369 1660Department of Radiology, Changhai Hospital of Shanghai, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433 China
| |
Collapse
|
27
|
Kamitani N, Nakamae I, Yoneda-Kato N, Kato JY, Sho M. Preclinical evaluation of pentagamavunone-1 as monotherapy and combination therapy for pancreatic cancer in multiple xenograft models. Sci Rep 2022; 12:22419. [PMID: 36575213 PMCID: PMC9794715 DOI: 10.1038/s41598-022-26863-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
We previously reported that pentagamavunone-1 (PGV-1) effectively inhibited cell proliferation in many types of human tumors, including pancreatic cancer, by inducing M phase (prometaphase) arrest, senescence, and apoptosis with few side effects. However, a detailed evaluation of the effects of PGV-1 on pancreatic cancer cells in an in vivo setting has not yet been conducted. The present study investigated the potential efficacy of PGV-1 as both monotherapy and combination therapy for pancreatic cancer using multiple xenograft mouse assays. A cell-line derived xenograft model (CDX-M) with pancreatic cancer cell line and a patient-derived xenograft mouse model (PDX-M) using resected pancreatic cancer samples without neoadjuvant chemotherapy were established in both heterotopic and orthotopic manners. PGV-1 effectively suppressed tumor formation at the heterotopic and orthotopic sites in CDX-M than in untreated mice. Combination therapy with PGV-1 and gemcitabine more effectively suppressed tumor formation than monotherapy with PGV-1 or gemcitabine when administered after tumor formation. Monotherapy with PGV-1 or gemcitabine less effectively suppressed tumor formation in PDX-M than in CDX-M, whereas combination therapy with PGV-1 and gemcitabine more effectively suppressed tumor formation. PGV-1 as monotherapy and combination therapy with gemcitabine effectively inhibited tumor formation and has potential as an anticancer candidate for pancreatic cancer.
Collapse
Affiliation(s)
- Naoki Kamitani
- grid.410814.80000 0004 0372 782XDepartment of Surgery, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara 634-8522 Japan
| | - Ikuko Nakamae
- grid.260493.a0000 0000 9227 2257Laboratory of Tumor Cell Biology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama, Ikoma, Nara 630-0101 Japan
| | - Noriko Yoneda-Kato
- grid.260493.a0000 0000 9227 2257Laboratory of Tumor Cell Biology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama, Ikoma, Nara 630-0101 Japan
| | - Jun-ya Kato
- grid.260493.a0000 0000 9227 2257Laboratory of Tumor Cell Biology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama, Ikoma, Nara 630-0101 Japan
| | - Masayuki Sho
- grid.410814.80000 0004 0372 782XDepartment of Surgery, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara 634-8522 Japan
| |
Collapse
|
28
|
Choi MH, Yoon SB. Sarcopenia in pancreatic cancer: Effect on patient outcomes. World J Gastrointest Oncol 2022; 14:2302-2312. [PMID: 36568942 PMCID: PMC9782618 DOI: 10.4251/wjgo.v14.i12.2302] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/29/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer is a challenging disease with an increasing incidence and extremely poor prognosis. The clinical outcomes of pancreatic cancer depend on tumor biology, responses to treatments, and malnutrition or cachexia. Sarcopenia represents a severe catabolic condition defined by the age-related loss of muscle mass and strength and affects as much as 70% of malnourished pancreatic cancer patients. The lumbar skeletal muscle index, defined as the total abdominal muscle area at the L3 vertebral level adjusted by the square of the height, is widely used for assessing sarcopenia in patients with pancreatic cancer. Several studies have suggested that sarcopenia may be a risk factor for perioperative complications and decreased recurrence-free or overall survival in patients with pancreatic cancer undergoing surgery. Sarcopenia could also intensify chemotherapy-induced toxicities and worsen the quality of life and survival in the neoadjuvant or palliative chemotherapy setting. Sarcopenia, not only at the time of diagnosis but also during treatment, decreases survival in patients with pancreatic cancer. Theoretically, multimodal interventions may improve sarcopenia and clinical outcomes; however, no study has reported positive results. Further prospective studies are needed to confirm the prognostic role of sarcopenia and the effects of multimodal interventions in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Moon Hyung Choi
- Department of Radiology, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 03312, South Korea
| | - Seung Bae Yoon
- Division of Gastroenterology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 03312, South Korea
| |
Collapse
|
29
|
Tseng HC, Kuo CY, Liao WT, Chou TS, Hsiao JK. Indocyanine green as a near-infrared theranostic agent for ferroptosis and apoptosis-based, photothermal, and photodynamic cancer therapy. Front Mol Biosci 2022; 9:1045885. [PMID: 36567945 PMCID: PMC9768228 DOI: 10.3389/fmolb.2022.1045885] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
Ferroptosis is a recently discovered programmed cell death pathway initiated by reactive oxygen species (ROS). Cancer cells can escape ferroptosis, and strategies to promote cancer treatment are crucial. Indocyanine green (ICG) is a near-infrared (NIR) fluorescent molecule used in the imaging of residual tumor removal during surgery. Growing attention has been paid to the anticancer potential of ICG-NIR irradiation by inducing ROS production and theranostic effects. Organic anion transmembrane polypeptide (OATP) 1B3 is responsible for ICG metabolism. Additionally, the overexpression of OATP1B3 has been reported in several cancers. However, whether ICG combined with NIR exposure can cause ferroptosis remains unknown and the concept of treating OATP1B3-expressing cells with ICG-NIR irradiation has not been validated. We then used ICG as a theranostic molecule and an OATP1B3-transfected fibrosarcoma cell line, HT-1080 (HT-1080-OATP1B3), as a cell model. The HT-1080-OATP1B3 cell could promote the uptake of ICG into the cytoplasm. We observed that the HT-1080-OATP1B3 cells treated with ICG and exposed to 808-nm laser irradiation underwent apoptosis, as indicated by a reduction in mitochondrial membrane potential, and upregulation of cleaved Caspase-3 and Bax but downregulation of Bcl-2 expression. Moreover, lipid ROS production and consequent ferroptosis and hyperthermic effect were noted after ICG and laser administration. Finally, in vivo study findings also revealed that ICG with 808-nm laser irradiation has a significant effect on cancer suppression. ICG is a theranostic molecule that exerts synchronous apoptosis, ferroptosis, and hyperthermia effects and thus can be used in cancer treatment. Our findings may facilitate the development of treatment modalities for chemo-resistant cancers.
Collapse
Affiliation(s)
- Hsiang-Ching Tseng
- Department of Medical Imaging, Taipei Tzu Chi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City, Taiwan,Department of Research, Taipei Tzu Chi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City, Taiwan
| | - Chan-Yen Kuo
- Department of Research, Taipei Tzu Chi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City, Taiwan
| | - Wei-Ting Liao
- Department of Research, Taipei Tzu Chi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City, Taiwan
| | - Te-Sen Chou
- Department of Medical Imaging, Taipei Tzu Chi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City, Taiwan,Department of Research, Taipei Tzu Chi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City, Taiwan
| | - Jong-Kai Hsiao
- Department of Medical Imaging, Taipei Tzu Chi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City, Taiwan,School of Medicine, Tzu Chi University, Hualien, Taiwan,*Correspondence: Jong-Kai Hsiao,
| |
Collapse
|
30
|
Wang B, Zhang K, Ding Y, Huang T. Diagnostic value of multi-slice spiral CT and MRI in peripheral lymph node metastasis of pancreatic cancer. Asian J Surg 2022; 46:2083-2084. [PMID: 36443205 DOI: 10.1016/j.asjsur.2022.11.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 11/11/2022] [Indexed: 11/27/2022] Open
|
31
|
Lu C, Zhu Y, Cheng H, Kong W, Zhu L, Wang L, Tang M, Chen J, Li Q, He J, Li A, Qiu X, Chen D, Meng F, Qian X, Liu B, Qiu Y, Du J. Case Report: Pathologic Complete Response to Induction Therapy in a Patient With Potentially Resectable Pancreatic Cancer. Front Oncol 2022; 12:898119. [PMID: 35734594 PMCID: PMC9207502 DOI: 10.3389/fonc.2022.898119] [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: 03/17/2022] [Accepted: 04/22/2022] [Indexed: 11/21/2022] Open
Abstract
Immune monotherapy does not appear to work in patients with pancreatic cancer so far. We are conducting a clinical trial that combines programmed cell death protein-1 (PD-1) inhibitor with chemotherapy and concurrent radiotherapy as induction therapy for patients with locally advanced pancreatic cancer (LAPC) and borderline resectable pancreatic cancer (BRPC). Here, we report a case with a pathologic complete response (pCR) and no postoperative complications after the induction therapy. The patient received four cycles of induction therapy and achieved a partial response (PR) with a significant decline of tumor marker carbohydrate antigen 19-9 (CA19-9). Also, peripheral blood samples were collected during the treatment to investigate serial circulating tumor DNA (ctDNA) dynamic changes in predicting the tumor response and outcomes in patients. Our result suggested that PD-1 blockade plus chemotherapy and concurrent radiotherapy is a promising mode as induction therapy for patients with potentially resectable pancreatic cancer. In this case, serial ctDNA alterations accurately provide a comprehensive outlook of the tumor status and monitor the response to the therapy, as validated by standard imaging.
Collapse
Affiliation(s)
- Changchang Lu
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China.,Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yahui Zhu
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Hao Cheng
- Department of Hepatopancreatobiliary Surgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Weiwei Kong
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Linxi Zhu
- Department of Hepatopancreatobiliary Surgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lei Wang
- Digestive Department of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Min Tang
- Imaging Department of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jun Chen
- Pathology Department of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qi Li
- Pathology Department of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jian He
- Nuclear Medicine Department of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Aimei Li
- Nuclear Medicine Department of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xin Qiu
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Dongsheng Chen
- The State Key Laboratory of Translational Medicine and Innovative Drug Development, Medical Department, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing, China
| | - Fanyan Meng
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiaoping Qian
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Baorui Liu
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yudong Qiu
- Department of Hepatopancreatobiliary Surgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Juan Du
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|