1
|
Son H, Lee J, Han SY, Kim TI, Kim DU, Kim D, Kim GH. Evaluation of cryoablation using a prototype cryoablation needle in swine liver. Clin Endosc 2024; 57:675-682. [PMID: 39354834 PMCID: PMC11474465 DOI: 10.5946/ce.2024.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/20/2024] [Accepted: 04/21/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND/AIMS Pancreatic cancer poses significant challenges due to its tendency for late-stage diagnosis and high mortality rates. Cryoablation, a technique used to treat various types of cancer, has shown potential in enhancing the prognosis of pancreatic cancer when combined with other therapies. However, its implementation is often limited by the need for lengthy procedures and specialized equipment. This study aims to develop a cryoablation needle optimized for endoscopic ultrasonography to simplify its application in treating pancreatic cancer. METHODS The study involved conducting cryoablation experiments on swine liver tissue. It utilized cryo-needles to evaluate the extent of cell death across various temperatures and durations of cryoablation. RESULTS The cryoablation system, which employed liquid carbon dioxide, achieved rapid cooling, reaching temperatures below -60 °C within 30 seconds and maintained the cryoablation process for 200 seconds. These conditions resulted in necrosis of the liver tissue. Notable cellular changes were observed up to 15 mm away from the cryoablation needle. CONCLUSIONS This experimental study successfully demonstrated the efficacy of using a cryo-needle for cryoablation in swine liver tissue. Further trials involving pancreatic tissue are expected to verify its effectiveness, underscoring the importance of continued research to establish its role as a complementary therapy in pancreatic cancer treatment.
Collapse
Affiliation(s)
- Hyunjoon Son
- Department of Mechanical Engineering, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Jonghyun Lee
- Division of Gastroenterology, Department of Internal Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Korea
| | - Sung Yong Han
- Division of Gastroenterology, Department of Internal Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Korea
| | - Tae In Kim
- Division of Gastroenterology, Department of Internal Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Korea
| | - Dong Uk Kim
- Department of Internal Medicine, CHA Gumi Medical Center, CHA University, Gumi, Korea
| | - Daejin Kim
- Department of Mechanical Engineering, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Gun-Ho Kim
- Department of Mechanical Engineering, Ulsan National Institute of Science and Technology, Ulsan, Korea
| |
Collapse
|
2
|
Son H, Kim TI, Lee J, Han SY, Kim DU, Kim D, Kim GH. A Preliminary Study of a Prototype Cryoablation Needle on Porcine Livers for Pancreatic Cancer Treatment. J Clin Med 2024; 13:4998. [PMID: 39274210 PMCID: PMC11396432 DOI: 10.3390/jcm13174998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 09/16/2024] Open
Abstract
Background and Aims: Despite its relatively low incidence rate compared to others, pancreatic cancer has a poor prognosis owing to its late detection and poor response to systemic chemotherapy. Because the effectiveness of chemotherapy is still restricted, the need for locoregional treatment is increasing. Cryoablation is an effective and minimally invasive treatment for some cancers, but its efficiency in pancreatic cancer is limited. Despite recent reports about promising outcomes, the optimal method and conditions of treatment are not known. In this preliminary study, we aimed to develop a cryoablation needle which can control the ablated area considering application through endoscopic ultrasonography. Methods: Here, we used a novel cryoneedle cooling system which can adjust the ablation range based on a liquid carbon dioxide refrigerant. Applied to the livers of swine, the cryoablation needle rapidly reached -60 °C within 30 s and cryoablation was performed for approximately 240 s. Based on the distance and depth, we collected real-time temperature data during the procedure. To compare the extent of cell death over time, tissue samples were collected hourly from 3 to 6 h after the procedure. Results: Approximately 4-5 mm of tissue was ablated via cryoablation, and cell death progressed over time after cryoablation. Moreover, the ablated lesions could be regulated using an insulating agent on the needle. Conclusions: This preliminary study on a novel surgical cooling needle system compatible with endoscopic ultrasound for cryoablation-based pancreatic cancer treatment confirmed the efficacy of cryoablation and identified the conditions necessary to induce necrosis. Additionally, this study evaluated the effectiveness of the insulation component of the system in protecting normal cells and assessed the extent of necrosis over time after the procedure.
Collapse
Affiliation(s)
- Hyunjoon Son
- Department of Mechanical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Tae In Kim
- Division of Gastroenterology, Department of Internal Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea
- Internal Medicine, School of Medicine, Pusan National University, Busan 46241, Republic of Korea
| | - Jonghyun Lee
- Division of Gastroenterology, Department of Internal Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea
- Internal Medicine, School of Medicine, Pusan National University, Busan 46241, Republic of Korea
| | - Sung Yong Han
- Division of Gastroenterology, Department of Internal Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea
- Internal Medicine, School of Medicine, Pusan National University, Busan 46241, Republic of Korea
| | - Dong Uk Kim
- Division of Gastroenterology, Department of Internal Medicine, CHA Gumi Medical Center, CHA University, Gumi 13488, Republic of Korea
| | - Daejin Kim
- Department of Mechanical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Gun-Ho Kim
- Department of Mechanical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| |
Collapse
|
3
|
Deng K, Zou F, Xu J, Xu D, Luo Z. Cancer-associated fibroblasts promote stemness maintenance and gemcitabine resistance via HIF-1α/miR-21 axis under hypoxic conditions in pancreatic cancer. Mol Carcinog 2024; 63:524-537. [PMID: 38197482 DOI: 10.1002/mc.23668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/24/2023] [Accepted: 11/24/2023] [Indexed: 01/11/2024]
Abstract
Gemcitabine (GEM) resistance affects chemotherapy efficacy of pancreatic cancer (PC). Cancer-associated fibroblasts (CAFs) possess the ability of regulating chemoresistance. This study probed the mechanism of hypoxia-treated CAFs regulating cell stemness and GEM resistance in PC. Miapaca-2/SW1990 were co-cultured with PC-derived CAFs under normoxic/hypoxic conditions. Cell viability/self-renewal ability was determined by MTT/sphere formation assays, respectively. Protein levels of CD44, CD133, Oct4, and Sox2 were determined by western blot. GEM tumoricidal assay was performed. PC cell GEM resistance was evaluated by MTT assay. CAFs were cultured at normoxia/hypoxia. HIF-1α and miR-21 expression levels were assessed by RT-qPCR and western blot, with their binding sites and binding relationship predicted and verified. CAF-extracellular vesicles (EVs) were incubated with Miapaca-2 cells. The RAS/AKT/ERK pathway activation was detected by western blot. PC xenograft models were established and treated with hypoxic CAF-EVs and GEM. CAFs and PC cell co-culture increased cell stemness maintenance, GEM resistance, cell viability, stem cell sphere number, and protein levels of CD44, CD133, Oct4, and Sox2, and weakened GEM tumoricidal ability to PC cells, with the effects further enhanced by hypoxia. Hypoxia induced HIF-1α and miR-21 overexpression in CAFs. Hypoxia promoted CAFs to secrete high-level miR-21 EVs via the HIF-1α/miR-21 axis, and activated the miR-21/RAS/AKT/ERK pathway. CAF-EVs promoted GEM resistance in PC via the miR-21/RAS/ATK/ERK pathway in vivo. Hypoxia promoted CAFs to secrete high-level miR-21 EVs through the HIF-1α/miR-21 axis, and activated the miR-21/RAS/AKT/ERK pathway via EVs to trigger stemness maintenance and GEM resistance in PC.
Collapse
Affiliation(s)
- Keping Deng
- Department of General Surgery, The First Hospital of Changsha (The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University), Changsha, Hunan Province, China
| | - Fang Zou
- Department of General Surgery, The First Hospital of Changsha (The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University), Changsha, Hunan Province, China
| | - Jin Xu
- Department of General Surgery, The First Hospital of Changsha (The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University), Changsha, Hunan Province, China
| | - Dayong Xu
- Department of General Surgery, The First Hospital of Changsha (The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University), Changsha, Hunan Province, China
| | - Zhen Luo
- Department of General Surgery, The First Hospital of Changsha (The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University), Changsha, Hunan Province, China
| |
Collapse
|
4
|
Baust JM, Robilotto A, Raijman I, Santucci KL, Van Buskirk RG, Baust JG, Snyder KK. The Assessment of a Novel Endoscopic Ultrasound-Compatible Cryocatheter to Ablate Pancreatic Cancer. Biomedicines 2024; 12:507. [PMID: 38540120 PMCID: PMC10968037 DOI: 10.3390/biomedicines12030507] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/15/2024] [Accepted: 02/21/2024] [Indexed: 11/11/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease that may be treated utilizing thermal therapies. Cryoablation is an effective, minimally invasive therapy that has been utilized for the treatment of various cancers, offering patients a quicker recovery and reduced side effects. Cryoablation has been utilized on a limited basis for the treatment of PDAC. With the recent reports on the success of cryoablation, there is a growing interest in the use of cryoablation as a standalone, minimally invasive procedure to treat PDAC. While offering a promising path, the application of cryoablation to PDAC is limited by current technologies. As such, there is a need for the development of new devices to support advanced treatment strategies for PDAC. To this end, this study investigated the performance of a new endoscopic ultrasound-compatible cryoablation catheter technology, FrostBite. We hypothesized that FrostBite would enable the rapid, effective, minimally invasive delivery of ultra-cold temperatures to target tissues, resulting in effective ablation via an endoscopic approach. Thermal properties and ablative efficacy were evaluated using a heat-loaded gel model, tissue-engineered models (TEMs), and an initial in vivo porcine study. Freeze protocols evaluated included single and repeat 3 and 5 min applications. Isotherm assessment revealed the generation of a 2.2 cm diameter frozen mass with the -20 °C isotherm reaching a diameter of 1.5 cm following a single 5 min freeze. TEM studies revealed the achievement of temperatures ≤ -20 °C at a diameter of 1.9 cm after a 5 min freeze. Fluorescent imaging conducted 24 h post-thaw demonstrated a uniformly shaped ellipsoidal ablative zone with a midline diameter of 2.5 cm, resulting in a total ablative volume of 6.9 cm3 after a single 5 min freeze. In vivo findings consistently demonstrated the generation of ablative areas measuring 2.03 cm × 3.2 cm. These studies demonstrate the potential of the FrostBite cryocatheter as an endoscopic ultrasound-based treatment option. The data suggest that FrostBite may provide for the rapid, effective, controllable freezing of cancerous pancreatic and liver tissues. This ablative power also offers the potential of improved safety margins via the minimally invasive nature of an endoscopic ultrasound-based approach or natural orifice transluminal endoscopic surgery (NOTES)-based approach. The results of this pre-clinical feasibility study show promise, affirming the need for further investigation into the potential of the FrostBite cryocatheter as an advanced, minimally invasive cryoablative technology.
Collapse
Affiliation(s)
- John M. Baust
- CPSI Biotech, Owego, NY 13827, USA
- Phase Therapeutics, Inc., Owego, NY 13827, USA
| | | | - Isaac Raijman
- Department of Medicine-Gastroenterology, Baylor College of Medicine, Houston, TX 77030, USA
- GI Alliance, Houston, TX 77030, USA
| | | | - Robert G. Van Buskirk
- CPSI Biotech, Owego, NY 13827, USA
- Center for Translational Stem Cell and Tissue Engineering, Binghamton University, Binghamton, NY 13902, USA
- Department of Biological Sciences, Binghamton University, Binghamton, NY 13902, USA
| | - John G. Baust
- Center for Translational Stem Cell and Tissue Engineering, Binghamton University, Binghamton, NY 13902, USA
- Department of Biological Sciences, Binghamton University, Binghamton, NY 13902, USA
| | - Kristi K. Snyder
- CPSI Biotech, Owego, NY 13827, USA
- Phase Therapeutics, Inc., Owego, NY 13827, USA
| |
Collapse
|
5
|
Shakhova M, Elagin V, Plekhanov A, Khilov A, Kurakina D, Kamensky V, Kirillin M. Post-Operational Photodynamic Therapy of the Tumor Bed: Comparative Analysis for Cold Knife and Laser Scalpel Resection. Biomedicines 2024; 12:291. [PMID: 38397893 PMCID: PMC11154242 DOI: 10.3390/biomedicines12020291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
In this paper, we report on a study regarding the efficiency of the post-operational phototherapy of the tumor bed after resection with both a cold knife and a laser scalpel in laboratory mice with CT-26 tumors. Post-operational processing included photodynamic therapy (PDT) with a topically applied chlorin-based photosensitizer (PS), performed at wavelengths of 405 or 660 nm, with a total dose of 150 J/cm2. The selected design of the tumor model yielded zero recurrence in the laser scalpel group and 92% recurrence in the cold knife group without post-processing, confirming the efficiency of the laser scalpel in oncology against the cold knife. The application of PDT after the cold knife resection decreased the recurrence rate to 70% and 42% for the 405 nm and 660 nm procedures, respectively. On the other hand, the application of PDT after the laser scalpel resection induced recurrence rates of 18% and 30%, respectively, for the considered PDT performance wavelengths. The control of the penetration of PS into the tumor bed by fluorescence confocal microscopy indicated the deeper penetration of PS in the case of the cold knife, which presumably provided deeper PDT action, while the low-dose light exposure of deeper tissues without PS, presumably, stimulated tumor recurrence, which was also confirmed by the differences in the recurrence rate in the 405 and 660 nm groups. Irradiation-only light exposures, in all cases, demonstrated higher recurrence rates compared to the corresponding PDT cases. Thus, the PDT processing of the tumor bed after resection could only be recommended for the cold knife treatment and not for the laser scalpel resection, where it could induce tumor recurrence.
Collapse
Affiliation(s)
- Maria Shakhova
- Department of Ear, Nose and Throat Diseases, FSBEI HE «Privolzhsky Research Medical University» MOH Russia, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603005, Russia;
| | - Vadim Elagin
- Institute of Experimental Oncology and Biomedical Technologies, FSBEI HE «Privolzhsky Research Medical University» MOH Russia, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603005, Russia; (A.P.); (V.K.)
| | - Anton Plekhanov
- Institute of Experimental Oncology and Biomedical Technologies, FSBEI HE «Privolzhsky Research Medical University» MOH Russia, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603005, Russia; (A.P.); (V.K.)
| | - Aleksandr Khilov
- A.V. Gaponov-Grekhov Institute of Applied Physics of the Russian Academy of Sciences, 46 Ulyanov St., Nizhny Novgorod 603155, Russia; (A.K.); (D.K.); (M.K.)
| | - Daria Kurakina
- A.V. Gaponov-Grekhov Institute of Applied Physics of the Russian Academy of Sciences, 46 Ulyanov St., Nizhny Novgorod 603155, Russia; (A.K.); (D.K.); (M.K.)
| | - Vladislav Kamensky
- Institute of Experimental Oncology and Biomedical Technologies, FSBEI HE «Privolzhsky Research Medical University» MOH Russia, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603005, Russia; (A.P.); (V.K.)
- A.V. Gaponov-Grekhov Institute of Applied Physics of the Russian Academy of Sciences, 46 Ulyanov St., Nizhny Novgorod 603155, Russia; (A.K.); (D.K.); (M.K.)
| | - Mikhail Kirillin
- A.V. Gaponov-Grekhov Institute of Applied Physics of the Russian Academy of Sciences, 46 Ulyanov St., Nizhny Novgorod 603155, Russia; (A.K.); (D.K.); (M.K.)
| |
Collapse
|
6
|
Gu C, Wang X, Wang K, Xie F, Chen L, Ji H, Sun J. Cryoablation triggers type I interferon-dependent antitumor immunity and potentiates immunotherapy efficacy in lung cancer. J Immunother Cancer 2024; 12:e008386. [PMID: 38272564 PMCID: PMC10824009 DOI: 10.1136/jitc-2023-008386] [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] [Accepted: 01/11/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Cryoablation is a minimally invasive option for patients with medically inoperable non-small cell lung cancer (NSCLC) and can trigger abscopal immune-regulatory effects. However, it remains unclear how cryoablation affects the host-level immune response in NSCLC. In this study, we investigated the local and systemic immunological effects of cryoablation and the potential of combining cryoablation with programmed cell death protein 1 (PD-1) blockade to boost immunotherapy efficacy in NSCLC. METHODS We first investigated systemic immunological effects induced by cryoablation in patients with early-stage NSCLC. Subsequently, we explored cryoablation-induced antitumor immunity and the underlying biological mechanisms using KP (Kras G12D/+, Tp53 -/-) mutant lung cancer cell allograft mouse models. Moreover, the synergistic efficacy of cryoablation and PD-1 blockade was explored in both mouse models and patients with unresectable NSCLC. RESULTS We found that cryoablation significantly increased circulating CD8+ T cell subpopulations and proinflammatory cytokines in patients with early-stage NSCLC. In lung cancer cell allograft mouse models, we demonstrated that cryoablation resulted in abscopal growth inhibition of contralateral, non-ablated tumors. Integrated analysis of bulk, single-cell RNA and T cell receptor (TCR) sequencing data revealed that cryoablation reprogrammed the intratumoral immune microenvironment and increased CD8+ T cell infiltration with higher effector signature, interferon (IFN) response, and cytolytic activity. Mechanistically, cryoablation promoted antitumor effect through the STING-dependent type I IFN signaling pathway, and type I IFN signaling blockade attenuated this antitumor effect. We also found that the combination of PD-1 blockade with cryoablation further inhibited tumor growth compared with either treatment alone in an allograft mouse model. Moreover, the combination therapy induced notable tumor suppression and CD8+ T cell infiltration in patients with unresectable NSCLC. CONCLUSIONS Our results provide mechanistic insights into how cryoablation triggers the antitumor immune effect in lung cancer, thereby potentiating programmed cell death ligand 1 (PD-L1)/PD-1 blockade efficacy in the clinical treatment of NSCLC.
Collapse
Affiliation(s)
- Chuanjia Gu
- Department of Respiratory Endoscopy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Respiratory Endoscopy, Shanghai, China
| | - Xue Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Kaiyu Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fangfang Xie
- Department of Respiratory Endoscopy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Respiratory Endoscopy, Shanghai, China
| | - Luonan Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, China
- Guangdong Institute of Intelligence Science and Technology, Hengqin, Zhuhai, Guangdong, China
| | - Hongbin Ji
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, China
| | - Jiayuan Sun
- Department of Respiratory Endoscopy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Respiratory Endoscopy, Shanghai, China
| |
Collapse
|
7
|
Guo R, Zheng P, Zhu S, Zeng Z, Li Z, Yang Y. Comprehensive analysis and identification of prognostic biomarkers and immunotherapeutic targets in the NADPH oxidase family (and its regulatory subunits) in pancreatic ductal adenocarcinoma. Clin Transl Oncol 2023; 25:3460-3470. [PMID: 37222951 DOI: 10.1007/s12094-023-03211-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/29/2023] [Indexed: 05/25/2023]
Abstract
PURPOSE This study aimed to evaluate the role of NADPH in pancreatic ductal adenocarcinoma using bioinformatic analyses and experimental validations. METHODS We compared the expression levels, performed GO and KEGG analysis of NADPH oxidase family and its regulatory subunits, and determined the survival of patients with pancreatic ductal adenocarcinoma by GEPIA, David and KM plotter. The relationship between their expression with immune infiltration levels, phagocytotic/NK cell immune checkpoints, recruitment-related molecules were detected by Timer 2.0 and TISIDB, respectively. Subsequently, their correlation with NK cell infiltration level was verified by immunohistochemistry. RESULTS The expression of some members of the NADPH oxidase family and its regulatory subunits was significantly increased in pancreatic ductal adenocarcinoma tissues compared to that in normal tissues and was positively correlated with natural killer (NK) cell infiltration. Furthermore, the NADPH oxidase family and its regulatory subunits were associated with survival and immune status in patients with pancreatic ductal adenocarcinoma, including chemokines, immune checkpoints, and immune infiltration levels of NK cells, monocytes, and myeloid-derived suppressor cells. CONCLUSIONS These results suggest the NADPH oxidase family and its regulatory subunits might serve as indicators for predicting the responsiveness to immunotherapy and outcome of patients with pancreatic ductal adenocarcinoma, providing a new perspective or strategy for immunotherapy in pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Ruiqi Guo
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China
- Department of Pathology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Panchun Zheng
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China
- Department of Pathology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shasha Zhu
- The Center for Clinical Molecular Medical Detection, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhen Zeng
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, China
| | - Zhenyu Li
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, China.
| | - Yaying Yang
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, China.
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China.
- Department of Pathology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
8
|
De Grandis MC, Ascenti V, Lanza C, Di Paolo G, Galassi B, Ierardi AM, Carrafiello G, Facciorusso A, Ghidini M. Locoregional Therapies and Remodeling of Tumor Microenvironment in Pancreatic Cancer. Int J Mol Sci 2023; 24:12681. [PMID: 37628865 PMCID: PMC10454061 DOI: 10.3390/ijms241612681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Despite the advances made in treatment, the prognosis of pancreatic ductal adenocarcinoma (PDAC) remains dismal, even in the locoregional and locally advanced stages, with high relapse rates after surgery. PDAC exhibits a chemoresistant and immunosuppressive phenotype, and the tumor microenvironment (TME) surrounding cancer cells actively participates in creating a stromal barrier to chemotherapy and an immunosuppressive environment. Recently, there has been an increasing use of interventional radiology techniques for the treatment of PDAC, although they do not represent a standard of care and are not included in clinical guidelines. Local approaches such as radiation therapy, hyperthermia, microwave or radiofrequency ablation, irreversible electroporation and high-intensity focused ultrasound exert their action on the tumor tissue, altering the composition and structure of TME and potentially enhancing the action of chemotherapy. Moreover, their action can increase antigen release and presentation with T-cell activation and reduction tumor-induced immune suppression. This review summarizes the current evidence on locoregional therapies in PDAC and their effect on remodeling TME to make it more susceptible to the action of antitumor agents.
Collapse
Affiliation(s)
| | - Velio Ascenti
- Postgraduate School of Diagnostic and Interventional Radiology, University of Milan, 20122 Milan, Italy; (V.A.); (C.L.)
| | - Carolina Lanza
- Postgraduate School of Diagnostic and Interventional Radiology, University of Milan, 20122 Milan, Italy; (V.A.); (C.L.)
| | - Giacomo Di Paolo
- Oncology Unit 1, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (M.C.D.G.); (G.D.P.)
| | - Barbara Galassi
- Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (B.G.); (M.G.)
| | - Anna Maria Ierardi
- Radiology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.M.I.); (G.C.)
| | - Gianpaolo Carrafiello
- Radiology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.M.I.); (G.C.)
- Department of Oncology and Haemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Antonio Facciorusso
- Section of Gastroenterology, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Michele Ghidini
- Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (B.G.); (M.G.)
| |
Collapse
|
9
|
Yılmaz S, Arıöz Habibi H. Percutaneous cryoablation of follicular thyroid carcinoma metastasis to the pancreas. Diagn Interv Radiol 2023; 29:167-169. [PMID: 36960584 PMCID: PMC10679586 DOI: 10.5152/dir.2022.21708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/13/2021] [Indexed: 01/15/2023]
Abstract
The following is a technical report of the successful cryoablation of pancreatic metastasis originating from follicular thyroid carcinoma. The patient was a 72-year-old female who underwent total thyroidectomy and radioiodine ablation for follicular carcinoma. One year after surgery, a positron emission tomography-computed tomography (PET-CT) examination, performed to demonstrate the source of the increased thyroglobulin, showed a fluorodeoxyglucose (FDG) avid mass located in the body of the pancreas. A percutaneous tru-cut biopsy was performed that revealed follicular thyroid carcinoma metastasis to the pancreas. Because of the patient's comorbidities, the patient underwent percutaneous cryoablation and made a successful recovery over the following 13 months. At the most recent follow-up, the thyroglobulin level was undetectable, and a PET-CT scan showed no FDG avid mass in the pancreas. To our knowledge, follicular carcinoma metastasis of the pancreas is extremely rare, and this is the first report of successful cryoablation of a metastatic tumor in the pancreas.
Collapse
Affiliation(s)
- Saim Yılmaz
- Department of Radiology, Private Varisson Radiology Center, Antalya, Turkey
| | | |
Collapse
|
10
|
Kuang G, Zhang Q, Jia J, Yu Y. Freezing biological organisms for biomedical applications. SMART MEDICINE 2022; 1:e20220034. [PMID: 39188743 PMCID: PMC11235656 DOI: 10.1002/smmd.20220034] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 12/07/2022] [Indexed: 08/28/2024]
Abstract
Biological organisms play important roles in human health, either in a commensal or pathogenic manner. Harnessing inactivated organisms or living organisms is a promising way to treat diseases. As two types of freezing, cryoablation makes it simple to inactivate organisms that must be in a non-pathogenic state when needed, while cryopreservation is a facile way to address the problem of long-term storage challenged by living organism-based therapy. In this review, we present the latest studies of freezing biological organisms for biomedical applications. To begin with, the freezing strategies of cryoablation and cryopreservation, as well as their corresponding technical essentials, are illustrated. Besides, biomedical applications of freezing biological organisms are presented, including transplantation, tissue regeneration, anti-infection therapy, and anti-tumor therapy. The challenges and prospects of freezing living organisms for biomedical applications are well discussed. We believe that the freezing method will provide a potential direction for the standardization and commercialization of inactivated or living organism-based therapeutic systems, and promote the clinical application of organism-based therapy.
Collapse
Affiliation(s)
- Gaizhen Kuang
- Pharmaceutical Sciences LaboratoryÅbo Akademi UniversityTurkuFinland
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouChina
| | - Qingfei Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouChina
| | - Jinxuan Jia
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouChina
| | - Yunru Yu
- Pharmaceutical Sciences LaboratoryÅbo Akademi UniversityTurkuFinland
| |
Collapse
|
11
|
Buscail L, Culetto A, Mokhrane FZ, Napoléon B, Meyrignac O, Molinier B, Lebrin M, Bournet B, Bérard E, Canivet C. Endoscopic ultrasound as a reliable tool for assessment of pancreatic adenocarcinoma treatment: Example of in situ gene therapy. Endosc Int Open 2022; 10:E910-E916. [PMID: 35692905 PMCID: PMC9187414 DOI: 10.1055/a-1799-7774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/17/2022] [Indexed: 11/29/2022] Open
Abstract
Background and study aims In pancreatic cancer, the antitumor effect can only be assessed by means of a computed tomography (CT) scan using RECIST (Response Evaluation Criteria in Solid Tumours) criteria. The aim of this study was to assess the intra-observer and interobserver agreement of endoscopic ultrasound (EUS) imaging in assessing tumor volume in primary pancreatic cancer. Patients and methods During a Phase 1 gene therapy trial, 21 patients had EUS before the first and second EUS-guided in situ gene therapy injections. All anonymized EUS files were then randomly distributed to three gastroenterologists/endosonographers and three radiologists (blind status). The largest tumor diameter was measured and the intraclass correlation coefficient (ICC) was determined. Results Intra-observer and interobserver agreements were good to excellent, regardless of operator experience (junior versus senior member of staff) (ICC: 0.65 to 0.84). A comparison of pretreatment and post-treatment measurements by the investigators highlighted a significant antitumor effect (-11 %; P = 0.0098), similar to that obtained during the generic protocol (-10 %; P = 0.0045). Conclusions Interobserver agreement regarding primary pancreatic adenocarcinoma measurements appears good to excellent, thus paving the way for the future inclusion of EUS assessments, particularly in trials assessing local therapies for pancreatic tumors.
Collapse
Affiliation(s)
- Louis Buscail
- Department of Gastroenterology and Pancreatology, CHU Toulouse-Rangueil (University Hospital Centre) and Toulouse University III, Toulouse, France,Centre for Clinical Investigation in Biotherapy, CHU Toulouse-Rangueil and INSERM U1436, Toulouse, France
| | - Adrian Culetto
- Department of Gastroenterology and Pancreatology, CHU Toulouse-Rangueil (University Hospital Centre) and Toulouse University III, Toulouse, France
| | - Fatima-Zhora Mokhrane
- Department of Radiology, CHU Toulouse-Rangueil and Toulouse University III, Toulouse, France
| | - Bertrand Napoléon
- Department of Gastroenterology, Jean Mermoz Hospital, Ramsay Générale de Santé (General Health), Lyon, France
| | - Olivier Meyrignac
- Department of Radiology, CHU Toulouse-Rangueil and Toulouse University III, Toulouse, France
| | - Baptiste Molinier
- Department of Radiology, CHU Toulouse-Rangueil and Toulouse University III, Toulouse, France
| | - Marine Lebrin
- Centre for Clinical Investigation in Biotherapy, CHU Toulouse-Rangueil and INSERM U1436, Toulouse, France
| | - Barbara Bournet
- Department of Gastroenterology and Pancreatology, CHU Toulouse-Rangueil (University Hospital Centre) and Toulouse University III, Toulouse, France
| | - Emilie Bérard
- Department of Epidemiology, CHU of Toulouse & UMR 1027, CERPOP, INSERM, UPS, Toulouse University, Toulouse, France
| | - Cindy Canivet
- Department of Gastroenterology and Pancreatology, CHU Toulouse-Rangueil (University Hospital Centre) and Toulouse University III, Toulouse, France
| |
Collapse
|
12
|
An In Vitro Investigation into Cryoablation and Adjunctive Cryoablation/Chemotherapy Combination Therapy for the Treatment of Pancreatic Cancer Using the PANC-1 Cell Line. Biomedicines 2022; 10:biomedicines10020450. [PMID: 35203660 PMCID: PMC8962332 DOI: 10.3390/biomedicines10020450] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 02/01/2023] Open
Abstract
As the incidence of pancreatic ductal adenocarcinoma (PDAC) continues to grow, so does the need for new strategies for treatment. One such area being evaluated is cryoablation. While promising, studies remain limited and questions surrounding basic dosing (minimal lethal temperature) coupled with technological issues associated with accessing PDAC tumors and tumor proximity to vasculature and bile ducts, among others, have limited the use of cryoablation. Additionally, as chemotherapy remains the first-line of attack for PDAC, there is limited information on the impact of combining freezing with chemotherapy. As such, this study investigated the in vitro response of a PDAC cell line to freezing, chemotherapy, and the combination of chemotherapy pre-treatment and freezing. PANC-1 cells and PANC-1 tumor models were exposed to cryoablation (freezing insult) and compared to non-frozen controls. Additionally, PANC-1 cells were exposed to varying sub-clinical doses of gemcitabine or oxaliplatin alone and in combination with freezing. The results show that freezing to −10 °C did not affect viability, whereas −15 °C and −20 °C resulted in a reduction in 1 day post-freeze viability to 85% and 20%, respectively, though both recovered to controls by day 7. A complete cell loss was found following a single freeze below −25 °C. The combination of 100 nM gemcitabine (1.1 mg/m2) pre-treatment and a single freeze at −15 °C resulted in near-complete cell death (<5% survival) over the 7-day assessment interval. The combination of 8.8 µM oxaliplatin (130 mg/m2) pre-treatment and a single −15 °C freeze resulted in a similar trend of increased PANC-1 cell death. In summary, these in vitro results suggest that freezing alone to temperatures in the range of −25 °C results in a high degree of PDAC destruction. Further, the data support a potential combinatorial chemo/cryo-therapeutic strategy for the treatment of PDAC. These results suggest that a reduction in chemotherapeutic dose may be possible when offered in combination with freezing for the treatment of PDAC.
Collapse
|
13
|
Hall WA, Erickson B, Crane CH. Evolving Concepts Regarding Radiation Therapy for Pancreatic Cancer. Surg Oncol Clin N Am 2021; 30:719-730. [PMID: 34511192 PMCID: PMC8462521 DOI: 10.1016/j.soc.2021.06.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
In todays practice most institutions individualize the use of adjuvant, neoadjuvant, and definitive RT based on their interpretation of the available data. This review highlights novel concepts and approaches to the use of RT that should be considered by the surgical oncologist.
Collapse
Affiliation(s)
- William A Hall
- Department of Radiation Oncology, Froedtert and the Medical College of Wisconsin, 8701 West Watertown Plank Road, Milwaukee, WI 53226, USA; Graduate School of Biomedical Sciences, Medical College of Wisconsin; Department of Surgery, Froedtert and the Medical College of Wisconsin.
| | - Beth Erickson
- Department of Radiation Oncology, Froedtert and the Medical College of Wisconsin, 8701 West Watertown Plank Road, Milwaukee, WI 53226, USA; Department of Surgery, Froedtert and the Medical College of Wisconsin
| | | |
Collapse
|
14
|
Sauchinone inhibits hypoxia-induced epithelial-mesenchymal transition in pancreatic ductal adenocarcinoma cells through the Wnt/β-catenin pathway. Anticancer Drugs 2021; 31:918-924. [PMID: 32889895 DOI: 10.1097/cad.0000000000000956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The hypoxic microenvironment is commonly found in various solid tumors including pancreatic ductal adenocarcinoma (PDAC). Saururus chinensis is a medicinal Chinese herb widely used because of documented anti-inflammatory and anti-angiogenic properties. Sauchinone is special active lignin extracted from S. chinensis and its biological functions have been extensively explored. Recent studies have found that sauchinone could affect tumor initiation, metastasis and progression of some cancers. However, the specific role of sauchinone in PDAC remains to be elucidated. The main aim of this study was to elucidate the involvement of sauchinone in the progression of PDAC under the hypoxic condition. The human PDAC cell lines PANC-1 and BxPC-3 were exposed to hypoxia and various concentrations of sauchinone. The CCK-8 assay was performed to detect cytotoxic effects of sauchinone on PDAC cells. The levels of vascular endothelial growth factor, hypoxia-inducible factor-1α, E-cadherin, N-cadherin, Wnt3a and β-catenin were examined by the western blot analysis. Wound healing and transwell assays were used to assess cell migration and invasion. The results showed that the migratory and invasive abilities of PDAC cells were enhanced after exposure to hypoxia and the expression of epithelial-mesenchymal transition markers was also significantly regulated by hypoxia. All these effects induced under the hypoxic condition were terminated by sauchinone treatment. In addition, sauchinone suppressed hypoxia-induced activation of the Wnt/β-catenin signaling pathway. Our study provided important insight into understanding the mechanisms of the anti-cancer effect of sauchinone. Taken together, we suggested that sauchinone may be considered a new therapeutic agent for PDAC treatment.
Collapse
|
15
|
Role of Curcumin in Regulating Long Noncoding RNA Expression in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1308:13-23. [PMID: 33861433 DOI: 10.1007/978-3-030-64872-5_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Phytochemicals are various compounds produced by plants. There is growing evidence on their potential health effects. Some of these compounds are considered as traditional medicines and used as painkillers, anti-inflammatory agents, and for other applications. One of these phytochemicals is curumin, a natural polyphenol derived from the turmeric plant (Curcuma longa L.). Curcumin is widely used as a food coloring, preservative and condiment. It has also been shown to have antioxidative and anti-inflammatory effects. Moreover, there is growing evidence that curcumin alters long noncoding RNAs (lncRNAs) in many kinds of cancer. These noncoding RNAs can cause epigenetic modulation in the expression of several genes. This study reviews reports of curcumin effects on lncRNAs in lung, prostate, colorectal, breast, pancreatic, renal, gastric, and ovarian cancers.
Collapse
|
16
|
Wu T, Jin T. Prevention and treatment of digestive tract issues induced by postoperative adjuvant chemotherapy for pancreatic cancer using Terra Flava Usta. Am J Transl Res 2021; 13:3182-3189. [PMID: 34017487 PMCID: PMC8129296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/11/2020] [Indexed: 06/12/2023]
Abstract
OBJECTIVE To investigate the effects and possible mechanism of prevention and treatment of digestive tract reactions to postoperative adjuvant chemotherapy for pancreatic cancer (PC) using terra flava usta. METHODS A total of 75 PC patients with postoperative adjuvant chemotherapy who were admitted to our hospital from July 2016 to August 2019 were selected and randomly divided into a control group (CG) (n=37) and an observation group (OG) (n=38). Thirty min before chemotherapy, the CG received 8 mg of ondansetron via an intravenous drip, while the OG received 8 mg of ondansetron via an intravenous drip and terra flava usta. The grades of acute nausea and vomiting and delayed nausea and vomiting were compared between the two groups. The levels of gastrin (GAS), motilin (MTL), vasoactive peptide (VIP), hemoglobin (Hb), prealbumin (PA), and albumin (ALB), the proportion of CD4+, CD8+ and NK cells in T cell subsets, and the levels of 5-hydroxytryptamine (5-HT) and substance P (SP) were compared between the two groups. RESULTS The grades of delayed nausea and vomiting in the OG were superior to those in the CG (P < 0.05). The OG had higher levels of GAS and MTL and lower level of VIP than the CG after chemotherapy (P < 0.05). After chemotherapy, the levels of Hb, PA and ALB in the OG were higher than those in the CG (P < 0.05). After chemotherapy, the proportion of CD4+ and NK cells in the OG were higher than those in the CG, while the proportion of CD8+ in the OG was lower than that in the CG (P < 0.05). The serum 5-HT level in the acute phase and the delayed phase was higher than that before chemotherapy (P < 0.05), and the serum 5-HT level in the delayed phase was lower than that in the acute phase (P < 0.05). There was no significant difference in serum 5-HT level between the two groups in the acute phase and the delayed phase (P > 0.05). Compared with that before chemotherapy, SP levels in the OG decreased in the acute phase and the delayed phase, while SP levels in the CG did not change significantly, and SP levels in the OG were lower than those in the CG (P < 0.05). CONCLUSION Terra flava usta can effectively prevent and treat digestive tract reactions (e.g., delayed nausea and vomiting) induced by postoperative adjuvant chemotherapy for PC and protect gastrointestinal function, nutritional status and immune functions of PC patients. The mechanism of prevention and treatment of delayed digestive tract reactions after chemotherapy with terra flava usta may be related to the regulation of SP content. Therefore, terra flava usta is worthy of promotion and implementation.
Collapse
Affiliation(s)
- Tingting Wu
- Department of Traditional Chinese Medicine, Central Hospital Affiliated to Shandong First Medical UniversityJi’nan 250013, Shandong, China
| | - Tao Jin
- Department of Pharmacy, Central Hospital Affiliated to Shandong First Medical UniversityJi’nan 250013, Shandong, China
| |
Collapse
|
17
|
Adil MS, Khulood D, Somanath PR. Targeting Akt-associated microRNAs for cancer therapeutics. Biochem Pharmacol 2020; 189:114384. [PMID: 33347867 DOI: 10.1016/j.bcp.2020.114384] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/19/2022]
Abstract
The uncontrolled growth and spread of abnormal cells because of activating protooncogenes and/or inactivating tumor suppressor genes are the hallmarks of cancer. The PI3K/Akt signaling is one of the most frequently activated pathways in cancer cells responsible for the regulation of cell survival and proliferation in stress and hypoxic conditions during oncogenesis. Non-coding RNAs are a large family of RNAs that are not involved in protein-coding, and microRNAs (miRNAs) are a sub-set of non-coding RNAs with a single strand of 18-25 nucleotides. miRNAs are extensively involved in the post-transcriptional regulation of gene expression and play an extensive role in the regulatory mechanisms including cell differentiation, proliferation, apoptosis, and tumorigenesis. The impact of cancer on mRNA stability and translation efficiency is extensive and therefore, cancerous tissues exhibit drastic alterations in the expression of miRNAs. miRNAs can be modulated by utilizing techniques such as miRNA mimics, miRNA antagonists, or CRISPR/Cas9. In addition to their capacity as potential targets in cancer therapy, they can be used as reliable biomarkers to diagnose the disease at the earliest stage. Recent evidence indicates that microRNA-mediated gene regulation intersects with the Akt pathway, forming an Akt-microRNA regulatory network. miRNAs and Akt in this network operate together to exert their cellular tasks. In the current review, we discuss the Akt-associated miRNAs in several cancers, their molecular regulation, and how this newly emerging knowledge may contribute greatly to revolutionize cancer therapy.
Collapse
Affiliation(s)
- Mir S Adil
- Clinical and Experimental Therapeutics, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, United States
| | - Daulat Khulood
- Clinical and Experimental Therapeutics, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, United States
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, United States.
| |
Collapse
|
18
|
Zheng Z, Zhao Y, An Y, Li N, Bo C, Ma W, Guo Y, Zou C, Tian B, Zhang S. Efficacy of argon-helium cryoablation and its effects on immune function of patients with neck malignant tumours. Clin Otolaryngol 2020; 46:206-212. [PMID: 32920994 DOI: 10.1111/coa.13648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/12/2020] [Accepted: 08/30/2020] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Our study aimed to investigate the clinical efficacy of argon-helium cryoablation and its effects on the immune function of patients with neck malignant tumours. DESIGN Retrospective study. SETTING Single-institution academic tertiary care centre. METHODS Totally, 180 patients harbouring head and neck malignant tumours were divided into the argon-helium cryoablation group (n = 150) and the radiotherapy group (n = 50). The efficacy of the two groups was compared, and the immune function was observed. RESULTS The short-term clinical effect of the argon-helium cryoablation group was significantly higher than that of the radiotherapy group (P < .05). After treatment, the CD3+, CD4+, CD8+ and CD4+/CD8+ of the argon-helium cryoablation group were significantly better than those of the radiotherapy group (P < .001). The results of TNF-α, IL-1 β and CRP in the argon-helium cryoablation group were significantly better than that in the radiotherapy group (P < .001). CONCLUSION Argon-helium cryoablation could effectively improve the immune function, 5-year survival rate and local remission rate.
Collapse
Affiliation(s)
- Zheng Zheng
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yingchun Zhao
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yonghui An
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Na Li
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Changwen Bo
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wenhua Ma
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ying Guo
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Changpeng Zou
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Bo Tian
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Sujing Zhang
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
19
|
Sarkar RR, Fero KE, Seible DM, Panjwani N, Matsuno RK, Murphy JD. A Population-Based Study of Morbidity After Pancreatic Cancer Diagnosis. J Natl Compr Canc Netw 2020; 17:432-440. [PMID: 31085756 DOI: 10.6004/jnccn.2018.7111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 12/10/2018] [Indexed: 11/17/2022]
Abstract
BACKGROUND Pancreatic cancer is an aggressive disease characterized by early and relentless tumor spread, thus leading healthcare providers to consider it a "distant disease." However, local pancreatic tumor progression can lead to substantial morbidity. This study defines the long-term morbidity from local and nonlocal disease progression in a large population-based cohort. METHODS A total of 21,500 Medicare beneficiaries diagnosed with pancreatic cancer in 2000 through 2011 were identified. Hospitalizations were attributed to complications of either local disease (eg, biliary disorder, upper gastrointestinal ulcer/bleed, pain, pancreas-related, radiation toxicity) or nonlocal/distant disease (eg, thromboembolic events, cytopenia, dehydration, nausea/vomiting/motility problem, malnutrition and cachexia, ascites, pathologic fracture, and chemotherapy-related toxicity). Competing risk analyses were used to identify predictors of hospitalization. RESULTS Of the total cohort, 9,347 patients (43.5%) were hospitalized for a local complication and 13,101 patients (60.9%) for a nonlocal complication. After adjusting for the competing risk of death, the 12-month cumulative incidence of hospitalization from local complications was highest in patients with unresectable disease (53.1%), followed by resectable (39.5%) and metastatic disease (33.7%) at diagnosis. For nonlocal complications, the 12-month cumulative incidence was highest in patients with metastatic disease (57.0%), followed by unresectable (56.8%) and resectable disease (42.8%) at diagnosis. Multivariable analysis demonstrated several predictors of hospitalization for local and nonlocal complications, including age, race/ethnicity, location of residence, disease stage, tumor size, and diagnosis year. Radiation and chemotherapy had minimal impact on the risk of hospitalization. CONCLUSIONS Despite the widely known predilection of nonlocal/distant disease spread in pancreatic cancer, local tumor progression also leads to substantial morbidity and frequent hospitalization.
Collapse
Affiliation(s)
- Reith R Sarkar
- Department of Radiation Medicine and Applied Sciences, UC San Diego Moores Cancer Center, La Jolla, California
| | - Katherine E Fero
- Department of Radiation Medicine and Applied Sciences, UC San Diego Moores Cancer Center, La Jolla, California
| | - Daniel M Seible
- Department of Radiation Medicine and Applied Sciences, UC San Diego Moores Cancer Center, La Jolla, California
| | - Neil Panjwani
- Department of Radiation Medicine and Applied Sciences, UC San Diego Moores Cancer Center, La Jolla, California
| | - Rayna K Matsuno
- Department of Radiation Medicine and Applied Sciences, UC San Diego Moores Cancer Center, La Jolla, California
| | | |
Collapse
|
20
|
Hu Y, Wang B, Wang L, Wang Z, Jian Z, Deng L. Mammalian STE20‑like kinase 1 regulates pancreatic cancer cell survival and migration through Mfn2‑mediated mitophagy. Mol Med Rep 2020; 22:398-404. [PMID: 32377725 PMCID: PMC7248474 DOI: 10.3892/mmr.2020.11098] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/19/2020] [Indexed: 12/11/2022] Open
Abstract
Mammalian STE20-like kinase 1 (MST1) plays an important role in pancreatic cancer progression, but its downstream targets are still unknown. In the present study, our results indicated that MST1 expression was significantly downregulated in pancreatic cancer cell lines (PANC‑1, BxPC‑3 and HPAC) compared with that in the normal ductal epithelial cell line (hTERT‑HPNE). Moreover, MST1 overexpression in PANC‑1 cells led to increased apoptosis as determined by MTT and TUNEL assays and inhibited cellular migration. Mechanistically, upregulation of MST1 expression caused mitochondrial dysfunction, decreased ATP production, and activation of the mitochondrial‑dependent apoptotic pathway via inhibition of mitofusin 2 (Mfn2)‑mediated mitophagy, which ultimately resulted in increased cellular apoptosis and decreased cellular migration. Collectively, the present study demonstrated that MST1 may regulate pancreatic cancer PANC‑1 cell survival, invasion and migration through Mfn2‑mediated mitophagy, laying the foundation for the exploration of novel therapeutic targets for pancreatic cancer.
Collapse
Affiliation(s)
- Yongli Hu
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Bing Wang
- Department of General Surgery, 900th Hospital of the Joint Service Support Force of the PLA, Fuzhou, Fujian 350000, P.R. China
| | - Lie Wang
- Department of General Surgery, 900th Hospital of the Joint Service Support Force of the PLA, Fuzhou, Fujian 350000, P.R. China
| | - Zhenran Wang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Zhiyuan Jian
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Lin Deng
- Department of General Surgery, 900th Hospital of the Joint Service Support Force of the PLA, Fuzhou, Fujian 350000, P.R. China
| |
Collapse
|
21
|
Hu X, Ke G, Jang SRJ. Modeling Pancreatic Cancer Dynamics with Immunotherapy. Bull Math Biol 2019; 81:1885-1915. [PMID: 30843136 DOI: 10.1007/s11538-019-00591-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 02/22/2019] [Indexed: 12/11/2022]
Abstract
We develop a mathematical model of pancreatic cancer that includes pancreatic cancer cells, pancreatic stellate cells, effector cells and tumor-promoting and tumor-suppressing cytokines to investigate the effects of immunotherapies on patient survival. The model is first validated using the survival data of two clinical trials. Local sensitivity analysis of the parameters indicates there exists a critical activation rate of pro-tumor cytokines beyond which the cancer can be eradicated if four adoptive transfers of immune cells are applied. Optimal control theory is explored as a potential tool for searching the best adoptive cellular immunotherapies. Combined immunotherapies between adoptive ex vivo expanded immune cells and TGF-[Formula: see text] inhibition by siRNA treatments are investigated. This study concludes that mono-immunotherapy is unlikely to control the pancreatic cancer and combined immunotherapies between anti-TGF-[Formula: see text] and adoptive transfers of immune cells can prolong patient survival. We show through numerical explorations that how these two types of immunotherapies are scheduled is important to survival. Applying TGF-[Formula: see text] inhibition first followed by adoptive immune cell transfers can yield better survival outcomes.
Collapse
Affiliation(s)
- Xiaochuan Hu
- Department of Mathematics and Statistics, Texas Tech University, Lubbock, TX, 79409-1042, USA.,Department of Mathematics and Statistics, University of the Incarnate Word, San Antonio, TX, 78209, USA
| | - Guoyi Ke
- Department of Mathematics and Statistics, Texas Tech University, Lubbock, TX, 79409-1042, USA.,Department of Mathematics and Physical Sciences, Louisiana State University of Alexandria, Alexandria, LA, 71302, USA
| | - Sophia R-J Jang
- Department of Mathematics and Statistics, Texas Tech University, Lubbock, TX, 79409-1042, USA.
| |
Collapse
|
22
|
Saccomandi P, Lapergola A, Longo F, Schena E, Quero G. Thermal ablation of pancreatic cancer: A systematic literature review of clinical practice and pre-clinical studies. Int J Hyperthermia 2018; 35:398-418. [PMID: 30428728 DOI: 10.1080/02656736.2018.1506165] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
PURPOSE Pancreatic cancer is a challenging malignancy with low treatment option and poor life expectancy. Thermal ablation techniques were proposed as alternative treatment options, especially in advanced stages and for unfit-for-surgery patients. This systematic review describes the thermal ablative techniques -i.e., Laser (LA), Radiofrequency (RFA), Microwave (MWA) Ablation, High-Intensity Focused Ultrasound (HIFU) and cryoablation- available for pancreatic cancer treatment. Additionally, an analysis of the efficacy, complication rate and overall survival for each technique is conducted. MATERIAL AND METHODS This review collects the ex vivo, preclinical and clinical studies presenting the use of thermal techniques in the pancreatic cancer treatment, searched up to March 2018 in PubMed and Medline. Abstracts, letters-to-the-editor, expert opinions, reviews and non-English language manuscripts were excluded. RESULTS Sixty-five papers were included. For the ex vivo and preclinical studies, there are: 12 records for LA, 8 for RFA, 0 for MWA, 6 for HIFU, 1 for cryoablation and 3 for hybrid techniques. For clinical studies, 1 paper for LA, 14 for RFA, 1 for MWA, 17 for HIFU, 1 for cryoablation and 1 for hybrid techniques. CONCLUSIONS Important technological advances are presented in ex vivo and preclinical studies, as the real-time thermometry, nanotechnology and hybrid techniques to enhance the thermal outcome. Conversely, a lack of standardization in the clinical employment of the procedures emerged, leading to contrasting results on the safety and feasibility of some analyzed techniques. Uniform conclusions on the safety and feasibility of these techniques for pancreatic cancer will require further structured investigation.
Collapse
Affiliation(s)
- Paola Saccomandi
- a IHU-Strasbourg Institute of Image-Guided Surgery , Strasbourg , France.,b Departement of Mechanical Engineering, Politecnico di Milano , Milan , Italy
| | - Alfonso Lapergola
- a IHU-Strasbourg Institute of Image-Guided Surgery , Strasbourg , France.,c Università G. D'Annunzio , Chieti , Italy
| | - Fabio Longo
- a IHU-Strasbourg Institute of Image-Guided Surgery , Strasbourg , France.,d Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Rome , Italy
| | | | - Giuseppe Quero
- d Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Rome , Italy
| |
Collapse
|
23
|
Abdo J, Cornell DL, Mittal SK, Agrawal DK. Immunotherapy Plus Cryotherapy: Potential Augmented Abscopal Effect for Advanced Cancers. Front Oncol 2018; 8:85. [PMID: 29644213 PMCID: PMC5882833 DOI: 10.3389/fonc.2018.00085] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/12/2018] [Indexed: 01/10/2023] Open
Abstract
Since the 1920s the gold standard for treating cancer has been surgery, which is typically preceded or followed with chemotherapy and/or radiation, a process that perhaps contributes to the destruction of a patient’s immune defense system. Cryosurgery ablation of a solid tumor is mechanistically similar to a vaccination where hundreds of unique antigens from a heterogeneous population of tumor cells derived from the invading cancer are released. However, releasing tumor-derived self-antigens into circulation may not be sufficient enough to overcome the checkpoint escape mechanisms some cancers have evolved to avoid immune responses. The potentiated immune response caused by blocking tumor checkpoints designed to prevent programmed cell death may be the optimal treatment method for the immune system to recognize these new circulating cryoablated self-antigens. Preclinical and clinical evidence exists for the complementary roles for Cytotoxic T-lymphocyte-associated protein (CTLA-4) and PD-1 antagonists in regulating adaptive immunity, demonstrating that combination immunotherapy followed by cryosurgery provides a more targeted immune response to distant lesions, a phenomenon known as the abscopal effect. We propose that when the host’s immune system has been “primed” with combined anti-CTLA-4 and anti-PD-1 adjuvants prior to cryosurgery, the preserved cryoablated tumor antigens will be presented and processed by the host’s immune system resulting in a robust cytotoxic CD8+ T-cell response. Based on recent investigations and well-described biochemical mechanisms presented herein, a polyvalent autoinoculation of many tumor-specific antigens, derived from a heterogeneous population of tumor cancer cells, would present to an unhindered yet pre-sensitized immune system yielding a superior advantage in locating, recognizing, and destroying tumor cells throughout the body.
Collapse
Affiliation(s)
- Joe Abdo
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States
| | - David L Cornell
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States.,Department of Surgery, CHI Health Creighton University Medical Center, Omaha, NE, United States
| | - Sumeet K Mittal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States.,Dignity Health, Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Devendra K Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States
| |
Collapse
|
24
|
|
25
|
Abstract
The vast majority of patients who present with pancreatic adenocarcinoma have locally advanced or metastatic disease at the time of presentation without possibility of cure. Although in recent years there have been some new promising chemotherapy regimens that improve overall survival by a few months, the prognosis remains dismal. There is, however, a subset of patients who experience durable stable disease or partial responses after initial courses of chemotherapy with locally advanced disease. In these select patients, there remains interest in local ablative therapy with or without resection as a means for local control, palliation of symptoms, and possible improved survival. This review describes the techniques, complications, and expected benefits of several ablative techniques as a treatment modality for locally advanced pancreatic cancer.
Collapse
Affiliation(s)
- Rupen Shah
- From the Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, NY
| | | | | | | |
Collapse
|
26
|
He L, Niu L, Korpan NN, Sumida S, Xiao Y, Li J, Sutedja B, Lu Y, Zuo J, Liu J, Xu K. Clinical Practice Guidelines for Cryosurgery of Pancreatic Cancer: A Consensus Statement From the China Cooperative Group of Cryosurgery on Pancreatic Cancer, International Society of Cryosurgery, and Asian Society of Cryosurgery. Pancreas 2017; 46:967-972. [PMID: 28742542 PMCID: PMC5555970 DOI: 10.1097/mpa.0000000000000878] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/08/2017] [Indexed: 12/12/2022]
Abstract
Pancreatic cancer (PC), one of the most lethal malignancies, accounts for 8% to 10% of digestive system cancers, and the incidence is increasing. Surgery, chemotherapy, and radiotherapy have been the main treatment methods but are not very effective. Cryosurgery was first used in 1984 for treatment of locally advanced PC and has since become a considerable treatment for most cases of unresectable PC. During the past decade, cryosurgery has been applied in some hospitals in China, and the newly developed technique of computed tomography- and/or ultrasound-guided percutaneous cryosurgery has shown better results than chemotherapy in cases of unresectable locally advanced PC, with the 1-year survival rate reported to be more than 50%. To develop standardized criteria for the application of cryosurgery in PC, the International Society of Cryosurgery and Asian Society of Cryosurgery assembled experts from Austria, Japan, and China to discuss treatment methods and arrive at a consensus on the indications, contraindications, and preferred techniques of PC cryosurgery.
Collapse
Affiliation(s)
- Lihua He
- From the *Department of Oncology, Fuda Cancer Hospital, Jinan University School of Medicine, Guangzhou, China; †International Society of Cryosurgery, Austria Society of Cryosurgery, International Institute for Cryosurgery, Vienna, Austria; ‡International Society of Cryosurgery, Japan Society for Low Temperature Medicine, Tokyo, Japan; §Asian Society of Cryosurgery; PLA General Hospital, Beijing; and ∥Oncological Intervention Department, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; ¶Indonesian Society of Cryosurgery, Jakarta, Indonesia; and #Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Lizhi Niu
- From the *Department of Oncology, Fuda Cancer Hospital, Jinan University School of Medicine, Guangzhou, China; †International Society of Cryosurgery, Austria Society of Cryosurgery, International Institute for Cryosurgery, Vienna, Austria; ‡International Society of Cryosurgery, Japan Society for Low Temperature Medicine, Tokyo, Japan; §Asian Society of Cryosurgery; PLA General Hospital, Beijing; and ∥Oncological Intervention Department, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; ¶Indonesian Society of Cryosurgery, Jakarta, Indonesia; and #Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Nikolai N. Korpan
- From the *Department of Oncology, Fuda Cancer Hospital, Jinan University School of Medicine, Guangzhou, China; †International Society of Cryosurgery, Austria Society of Cryosurgery, International Institute for Cryosurgery, Vienna, Austria; ‡International Society of Cryosurgery, Japan Society for Low Temperature Medicine, Tokyo, Japan; §Asian Society of Cryosurgery; PLA General Hospital, Beijing; and ∥Oncological Intervention Department, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; ¶Indonesian Society of Cryosurgery, Jakarta, Indonesia; and #Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Sajio Sumida
- From the *Department of Oncology, Fuda Cancer Hospital, Jinan University School of Medicine, Guangzhou, China; †International Society of Cryosurgery, Austria Society of Cryosurgery, International Institute for Cryosurgery, Vienna, Austria; ‡International Society of Cryosurgery, Japan Society for Low Temperature Medicine, Tokyo, Japan; §Asian Society of Cryosurgery; PLA General Hospital, Beijing; and ∥Oncological Intervention Department, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; ¶Indonesian Society of Cryosurgery, Jakarta, Indonesia; and #Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yueyong Xiao
- From the *Department of Oncology, Fuda Cancer Hospital, Jinan University School of Medicine, Guangzhou, China; †International Society of Cryosurgery, Austria Society of Cryosurgery, International Institute for Cryosurgery, Vienna, Austria; ‡International Society of Cryosurgery, Japan Society for Low Temperature Medicine, Tokyo, Japan; §Asian Society of Cryosurgery; PLA General Hospital, Beijing; and ∥Oncological Intervention Department, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; ¶Indonesian Society of Cryosurgery, Jakarta, Indonesia; and #Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jiaping Li
- From the *Department of Oncology, Fuda Cancer Hospital, Jinan University School of Medicine, Guangzhou, China; †International Society of Cryosurgery, Austria Society of Cryosurgery, International Institute for Cryosurgery, Vienna, Austria; ‡International Society of Cryosurgery, Japan Society for Low Temperature Medicine, Tokyo, Japan; §Asian Society of Cryosurgery; PLA General Hospital, Beijing; and ∥Oncological Intervention Department, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; ¶Indonesian Society of Cryosurgery, Jakarta, Indonesia; and #Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Barlian Sutedja
- From the *Department of Oncology, Fuda Cancer Hospital, Jinan University School of Medicine, Guangzhou, China; †International Society of Cryosurgery, Austria Society of Cryosurgery, International Institute for Cryosurgery, Vienna, Austria; ‡International Society of Cryosurgery, Japan Society for Low Temperature Medicine, Tokyo, Japan; §Asian Society of Cryosurgery; PLA General Hospital, Beijing; and ∥Oncological Intervention Department, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; ¶Indonesian Society of Cryosurgery, Jakarta, Indonesia; and #Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Youyong Lu
- From the *Department of Oncology, Fuda Cancer Hospital, Jinan University School of Medicine, Guangzhou, China; †International Society of Cryosurgery, Austria Society of Cryosurgery, International Institute for Cryosurgery, Vienna, Austria; ‡International Society of Cryosurgery, Japan Society for Low Temperature Medicine, Tokyo, Japan; §Asian Society of Cryosurgery; PLA General Hospital, Beijing; and ∥Oncological Intervention Department, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; ¶Indonesian Society of Cryosurgery, Jakarta, Indonesia; and #Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jiansheng Zuo
- From the *Department of Oncology, Fuda Cancer Hospital, Jinan University School of Medicine, Guangzhou, China; †International Society of Cryosurgery, Austria Society of Cryosurgery, International Institute for Cryosurgery, Vienna, Austria; ‡International Society of Cryosurgery, Japan Society for Low Temperature Medicine, Tokyo, Japan; §Asian Society of Cryosurgery; PLA General Hospital, Beijing; and ∥Oncological Intervention Department, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; ¶Indonesian Society of Cryosurgery, Jakarta, Indonesia; and #Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jianguo Liu
- From the *Department of Oncology, Fuda Cancer Hospital, Jinan University School of Medicine, Guangzhou, China; †International Society of Cryosurgery, Austria Society of Cryosurgery, International Institute for Cryosurgery, Vienna, Austria; ‡International Society of Cryosurgery, Japan Society for Low Temperature Medicine, Tokyo, Japan; §Asian Society of Cryosurgery; PLA General Hospital, Beijing; and ∥Oncological Intervention Department, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; ¶Indonesian Society of Cryosurgery, Jakarta, Indonesia; and #Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Kecheng Xu
- From the *Department of Oncology, Fuda Cancer Hospital, Jinan University School of Medicine, Guangzhou, China; †International Society of Cryosurgery, Austria Society of Cryosurgery, International Institute for Cryosurgery, Vienna, Austria; ‡International Society of Cryosurgery, Japan Society for Low Temperature Medicine, Tokyo, Japan; §Asian Society of Cryosurgery; PLA General Hospital, Beijing; and ∥Oncological Intervention Department, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; ¶Indonesian Society of Cryosurgery, Jakarta, Indonesia; and #Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
27
|
Wang C, Feng Z, Jiang K, Zuo X. [ARTICLE WITHDRAWN] Upregulation of MicroRNA-935 Promotes the Malignant Behaviors of Pancreatic Carcinoma PANC-1 Cells via Targeting Inositol Polyphosphate 4-Phosphatase Type I Gene (INPP4A). Oncol Res 2017; 25:559-569. [PMID: 27733216 PMCID: PMC7841058 DOI: 10.3727/096504016x14759554689565] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
THIS ARTICLE WAS WITHDRAWN BY THE PUBLISHER IN NOVEMBER 2020
Collapse
Affiliation(s)
- Cuiyue Wang
- *Department of Digestive Disease, Qilu Hospital of Shandong University, Jinan, Shandong, P.R. China
- †Department of Digestive Disease, Linyi People’s Hospital, Linyi, Shandong, P.R. China
| | - Zhen Feng
- ‡Department of Joint Surgery, Linyi People’s Hospital, Linyi, Shandong, P.R. China
| | - Kaitong Jiang
- †Department of Digestive Disease, Linyi People’s Hospital, Linyi, Shandong, P.R. China
| | - Xiuli Zuo
- *Department of Digestive Disease, Qilu Hospital of Shandong University, Jinan, Shandong, P.R. China
| |
Collapse
|