1
|
Tsukada A, Miyajima I, Uchiyama S, Tabe D, Ikewaki N, Ichiyama K, Senthilkumar R, Iwasaki M, Abraham SJK. Effects of Nichi BRITE β-glucans as an onco-nutrition adjuvant in patients undergoing subtotal stomach-preserving pancreaticoduodenectomy (SSPPD) for pancreatic, bile duct and duodenal malignancies: A clinical study. Nutrition 2025; 137:112811. [PMID: 40383031 DOI: 10.1016/j.nut.2025.112811] [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: 06/28/2024] [Revised: 03/30/2025] [Accepted: 04/12/2025] [Indexed: 05/20/2025]
Abstract
OBJECTIVES Pancreatic cancers involve several challenges from difficulty in early diagnosis to high recurrence and mortality even after therapeutic interventions. Patients who undergo a surgical resection have several postoperative complications besides recurrence. Immunosuppression in the peri-operative period is a major challenge to overcome and in this study we investigated the effects of AFO-202 strain Aureobasidium-pullulans produced β-glucan (Nichi BRITE) in patients who underwent subtotal stomach-preserving pancreaticoduodenectomy (SSPPD) for pancreatic, bile duct and duodenal malignancies. METHODS Thirty patients undergoing SSPPD were randomly assigned to a control (n = 15) group. A placebo was given, and the treatment arm (n = 15), administered Nichi BRITE 250 mg (Glucan) in 3 divided doses per day, starting from 1 d prior to the day of surgery up to the 21st postoperative day, either by oral intake or feeding gastrostomy. Parameters of relevance to immune system and biomarkers of cancer pathology were evaluated. RESULTS Among the 22 patients who completed the study, the Nichi BRITE group showed notable increases in BG-IgA (+109.09%), CD209 (+54.68%, P = 0.034), and SAA (+800.70%, P = 0.050), while CA 19-9 decreased significantly (-5.86 U/mL, P < 0.001). Decrease in CD44 levels was greater in Nichi BRITE (-35.51%) than in the Placebo group (-14.05%). Disease-free survival (DFS) was longer in the Nichi BRITE group (16.1 mo) compared to the Placebo group (12.4 mo), with a lower recurrence rate (50% versus 71.4%) for pancreatic cancers. CONCLUSIONS Administration of Nichi BRITE β-glucan during the perioperative period in patients undergoing surgery for pancreatic, bile duct and duodenal malignancies has been safe, and it has yielded immune enhancement and improvement in biomarkers of better prognosis. Reduction in circulating cancer stem cells and pancreatic cancer marker CA19-9 inclines us to recommend this Nichi BRITE β-glucan be included in onco-nutrition guidelines for patients undergoing surgical removal of malignant tumors.
Collapse
Affiliation(s)
- Akira Tsukada
- Department of Surgical Gastroenterology, Chikamori Hospital, Kochi, Japan
| | - Isao Miyajima
- Department of Clinical Nutrition, Chikamori Hospital, Kochi, Japan
| | - Satomi Uchiyama
- Department of Clinical Nutrition, Chikamori Hospital, Kochi, Japan
| | - Daiki Tabe
- Department of Clinical Nutrition, Chikamori Hospital, Kochi, Japan
| | - Nobunao Ikewaki
- Department of Medical Life Science, Kyushu University of Medical Sciences, Japan; Institute of Immunology, Junsei Educational Institute, Nobeoka, Miyazaki, Japan
| | - Koji Ichiyama
- Antony- Xavier Interdisciplinary Scholastics (AXIS), GN Corporation Co. Ltd., Kofu, Japan
| | - Rajappa Senthilkumar
- Antony- Xavier Interdisciplinary Scholastics (AXIS), GN Corporation Co. Ltd., Kofu, Japan
| | - Masaru Iwasaki
- Centre for Advancing Clinical Research (CACR), University of Yamanashi - School of Medicine, Chuo, Japan
| | - Samuel J K Abraham
- Antony- Xavier Interdisciplinary Scholastics (AXIS), GN Corporation Co. Ltd., Kofu, Japan; Centre for Advancing Clinical Research (CACR), University of Yamanashi - School of Medicine, Chuo, Japan; Mary-Yoshio Translational Hexagon (MYTH), Nichi-In Centre for Regenerative Medicine (NCRM), Chennai, India; Levy-Jurgen Transdisciplinary Exploratory (LJTE), Global Niche Corp, Wilmington, Delaware, USA; R & D, Sophy Inc., Kochi, Japan; Haraguchi-Parikumar Advanced Remedies (HARP), SoulSynergy Ltd., Phoenix, Mauritius.
| |
Collapse
|
2
|
Qazi S, Trieu V. TGFB2 mRNA Levels Prognostically Interact with Interferon-Alpha Receptor Activation of IRF9 and IFI27, and an Immune Checkpoint LGALS9 to Impact Overall Survival in Pancreatic Ductal Adenocarcinoma. Int J Mol Sci 2024; 25:11221. [PMID: 39457004 PMCID: PMC11508538 DOI: 10.3390/ijms252011221] [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: 08/23/2024] [Revised: 09/13/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
The treatment of pancreatic ductal adenocarcinoma (PDAC) is an unmet challenge, with the median overall survival rate remaining less than a year, even with the use of FOLFIRINOX-based therapies. This study analyzed archived macrophage-associated mRNA expression using datasets deposited in the UCSC Xena web platform to compare normal pancreatic tissue and PDAC tumor samples. The TGFB2 gene exhibited low mRNA expression levels in normal tissue, with less than one TPM. In contrast, in tumor tissue, TGFB2 expression levels exhibited a 7.9-fold increase in mRNA expression relative to normal tissue (p < 0.0001). Additionally, components of the type-I interferon signaling pathway exhibited significant upregulation of mRNA levels in tumor tissue, including Interferon alpha/beta receptor 1 (IFNAR1; 3.4-fold increase, p < 0.0001), Interferon regulatory factor 9 (IRF9; 4.2-fold increase, p < 0.0001), Signal transducer and activator of transcription 1 (STAT1; 7.1-fold increase, p < 0.0001), and Interferon Alpha Inducible Protein 27 (IFI27; 66.3-fold increase, p < 0.0001). We also utilized TCGA datasets deposited in cBioportal and KMplotter to relate mRNA expression levels to overall survival outcomes. These increased levels of mRNA expression were found to be prognostically significant, whereby patients with high expression levels of either TGFB2, IRF9, or IFI27 showed median OS times ranging from 16 to 20 months (p < 0.01 compared to 72 months for patients with low levels of expression for both TGFB2 and either IRF9 or IFI27). Examination of the KMplotter database determined the prognostic impact of TGFB2 mRNA expression levels by comparing patients expressing high versus low levels of TGFB2 (50th percentile cut-off) in low macrophage TME. In TME with low macrophage levels, patients with high levels of TGFB2 mRNA exhibited significantly shorter OS outcomes than patients with low TGFB2 mRNA levels (Median OS of 15.3 versus 72.7 months, p < 0.0001). Furthermore, multivariate Cox regression models were applied to control for age at diagnosis. Nine genes exhibited significant increases in hazard ratios for TGFB2 mRNA expression, marker gene mRNA expression, and a significant interaction term between TGFB2 and marker gene expression (mRNA for markers: C1QA, CD74, HLA-DQB1, HLA-DRB1, HLA-F, IFI27, IRF9, LGALS9, MARCO). The results of our study suggest that a combination of pharmacological tools can be used in treating PDAC patients, targeting both TGFB2 and the components of the type-I interferon signaling pathway. The significant statistical interaction between TGFB2 and the nine marker genes suggests that TGFB2 is a negative prognostic indicator at low levels of the IFN-I activated genes and TAM marker expression, including the immune checkpoint LGALS9 (upregulated 16.5-fold in tumor tissue; p < 0.0001).
Collapse
MESH Headings
- Humans
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/mortality
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/mortality
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Prognosis
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Female
- Male
- Gene Expression Regulation, Neoplastic
- Transforming Growth Factor beta2/genetics
- Transforming Growth Factor beta2/metabolism
- Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics
- Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism
- Middle Aged
- Aged
- Receptor, Interferon alpha-beta/genetics
- Receptor, Interferon alpha-beta/metabolism
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Galectins
Collapse
Affiliation(s)
- Sanjive Qazi
- Oncotelic Therapeutics, 29397 Agoura Road, Suite 107, Agoura Hills, CA 91301, USA;
| | | |
Collapse
|
3
|
Sarfraz Z, Sarfraz A, Farooq MD, Khalid M, Cheema K, Javad F, Khan T, Pervaiz Z, Sarfraz M, Jaan A, Sadiq S, Anwar J. The Current Landscape of Clinical Trials for Immunotherapy in Pancreatic Cancer: A State-of-the-Art Review. J Gastrointest Cancer 2024; 55:1026-1057. [PMID: 38976079 DOI: 10.1007/s12029-024-01078-8] [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] [Accepted: 06/09/2024] [Indexed: 07/09/2024]
Abstract
BACKGROUND Pancreatic cancer remains a lethal malignancy with a 5-year survival rate below 6% and about 500,000 deaths annually worldwide. Pancreatic adenocarcinoma, the most prevalent form, is commonly associated with diabetes, chronic pancreatitis, obesity, and smoking, mainly affecting individuals aged 60 to 80 years. This systematic review aims to evaluate the efficacy of immunotherapeutic approaches in the treatment of pancreatic cancer. METHODS A systematic search was conducted to identify clinical trials (Phases I-III) assessing immunotherapy in pancreatic cancer in PubMed/Medline, CINAHL, Scopus, and Web of Science, adhering to PRISMA Statement 2020 guidelines. The final search was completed on May 25, 2024. Ongoing trials were sourced from ClinicalTrials.gov and the World Health Organization's International Clinical Trials Registry Platform (ICTRP). Keywords such as "pancreatic," "immunotherapy," "cancer," and "clinical trial" were used across databases. Gray literature was excluded. RESULTS Phase I trials, involving 337 patients, reported a median overall survival (OS) of 13.6 months (IQR: 5-62.5 months) and a median progression-free survival (PFS) of 5.1 months (IQR: 1.9-11.7 months). Phase II/III trials pooled in a total of 1463 participants had a median OS of 12.2 months (IQR: 2.5-35.55 months) and a median PFS of 8.8 months (IQR: 1.4-33.51 months). CONCLUSIONS Immunotherapy shows potential for extending survival among pancreatic cancer patients, though results vary. The immunosuppressive nature of the tumor microenvironment and diverse patient responses underline the need for further research to optimize these therapeutic strategies.
Collapse
Affiliation(s)
- Zouina Sarfraz
- Department of Medicine, Fatima Jinnah Medical University, Queen's Road, Mozang Chungi, Lahore, Pakistan.
| | | | | | - Musfira Khalid
- Department of Medicine, Fatima Jinnah Medical University, Queen's Road, Mozang Chungi, Lahore, Pakistan
| | | | | | - Taleah Khan
- CMH Lahore Medical College and Institute of Dentistry, Lahore, Pakistan
| | - Zainab Pervaiz
- CMH Lahore Medical College and Institute of Dentistry, Lahore, Pakistan
| | | | - Ali Jaan
- Rochester General Hospital, Rochester, NY, USA
| | | | - Junaid Anwar
- Baptist Hospitals of Southeast Texas, Beaumont, TX, USA
| |
Collapse
|
4
|
He Y, Kong L, Ji X, Zhuo M, An T, Jia B, Chi Y, Wang J, Zhao J, Li J, Yang X, Chen H, Zhai X, Tai Y, Ding L, Wang Z, Wang Y. Women patients with small-cell lung cancer using immunotherapy in a real-world cohort achieved long-term survival. Thorac Cancer 2024; 15:1727-1738. [PMID: 38923348 PMCID: PMC11320081 DOI: 10.1111/1759-7714.15393] [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: 04/12/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Usage of immune checkpoint inhibitors (ICIs) has prolonged the overall survival (OS) of patients with extensive-stage small-cell lung cancer (ES-SCLC). In clinical trials, males accounted for a large proportion, leading to the uncertainty of its efficacy in female patients. We therefore conducted this study to explore the efficacy and safety of using ICIs in female patients with ES-SCLC. METHODS We retrospectively enrolled female SCLC patients and subdivided them into two groups. Group A (n = 40) was defined as ES-SCLC patients who received first-line standard chemotherapy with or without ICIs. Group B (n = 47) included relapsed SCLC patients who were administered with second-line therapies. Kaplan-Meier methodology was used to calculate survival analysis. Chi-squared tests were used to analyze the incidence of adverse events (AEs). RESULTS Median progression-free survival (PFS) and median OS favored the ICI-contained cohorts (Group A PFS: 8.3 vs. 6.1 months; OS: not reached vs. 11.3 months; Group B PFS: 15.1 vs. 3.3 months; OS: 35.3 vs. 8.3 months), especially in those patients who received second-line immunotherapies. Patients who received immunotherapy had a slightly higher incidence rate of grade ≥3 AEs (Group A: 71.4% vs. 46.2%; Group B: 44.5% vs. 13.2%). Those who developed grade ≥3 AEs in first-line ICIs cohort had a more favorable survival (PFS: 8.3 vs. 3.2 months; OS: not reached vs. 5.1 months). CONCLUSIONS Our study suggested that female ES-SCLC patients treated with immunotherapy tended to achieve a relatively longer survival. The incidence of AEs (grade ≥3) was higher in women patients receiving ICIs, which requires monitoring more closely.
Collapse
Affiliation(s)
- Yuling He
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Lingdong Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Xumeng Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Minglei Zhuo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Tongtong An
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Bo Jia
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Yujia Chi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Jingjing Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Jun Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Jianjie Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Xue Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Hanxiao Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Xiaoyu Zhai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Yidi Tai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Lu Ding
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Ziping Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Yuyan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| |
Collapse
|
5
|
Quilbe A, Mustapha R, Duchêne B, Kumar A, Werkmeister E, Leteurtre E, Moralès O, Jonckheere N, Van Seuningen I, Delhem N. A novel anti-galectin-9 immunotherapy limits the early progression of pancreatic neoplastic lesions in transgenic mice. Front Immunol 2023; 14:1267279. [PMID: 38098486 PMCID: PMC10720041 DOI: 10.3389/fimmu.2023.1267279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/10/2023] [Indexed: 12/17/2023] Open
Abstract
Background Pancreatic adenocarcinoma (PDAC) is a devastating disease with an urgent need for therapeutic innovation. Immune checkpoint inhibition has shown promise in a variety of solid tumors, but most clinical trials have failed to demonstrate clinical efficacy in PDAC. This low efficacy is partly explained by a highly immunosuppressive microenvironment, which dampens anti-tumor immunity through the recruitment or induction of immunosuppressive cells, particularly regulatory T cells (Tregs). In this context, our laboratory has developed a novel immunotherapeutic strategy aimed at inhibiting the suppressive activity of Tregs, based on a patented (EP3152234B1) monoclonal antibody (mAb) targeting galectin-9 (LGALS9). Materials and methods CD4+ conventional T cells (TCD4 or Tconv), Treg ratio, and LGALS9 expression were analyzed by immunohistochemistry (IHC) and cytometry in blood and pancreas of K-rasLSL.G12D/+;Pdx-1-Cre (KC) and K-rasWildType (WT);Pdx1-Cre (WT) mice aged 4-13 months. Pancreatic intraepithelial neoplasm (PanIN) progression and grade were quantified using FIJI software and validated by pathologists. The anti-galectin-9 mAb was validated for its use in mice on isolated murine C57BL/6 Treg by immunofluorescence staining and cytometry. Its specificity and functionality were validated in proliferation assays on rLGALS9-immunosuppressed murine Tconv and in suppression assays between murine Treg and Tconv. Finally, 2-month-old KC mice were treated with anti-LGALS9 and compared to WT mice for peripheral and infiltrating TCD4, Treg, and PanIN progression. Results IHC and cytometry revealed a significant increase in LGALS9 expression and Treg levels in the blood and pancreas of KC mice proportional to the stages of precancerous lesions. Although present in WT mice, LGALS9 is expressed at a basal level with low and restricted expression that increases slightly over time, while Treg cells are few in number in their circulation and even absent from the pancreas over time. Using our anti-LGALS9 mAb in mice, it is shown that (i) murine Treg express LGALS9, (ii) the mAb could target and inhibit recombinant murine LGALS9, and (iii) neutralize murine Treg suppressive activity. Finally, the anti-LGALS9 mAb in KC mice reduced (i) LGALS9 expression in pancreatic cancer cells, (ii) the Treg ratio, and (iii) the total surface area and grade of PanIN. Conclusion We demonstrate for the first time that an anti-LGALS9 antibody, by specifically targeting endogenous LGALS9 tumor and exogenous LGALS9 produced by Treg, was able to limit the progression of pancreatic neoplastic lesions in mice, opening up new prospects for its use as an immunotherapeutic tool in PDAC.
Collapse
Affiliation(s)
- Alexandre Quilbe
- Univ. Lille, Inserm, CHU Lille, U1189 - ONCO-THAI - Assisted Laser Therapy and Immunotherapy for Oncology, Lille, France
| | - Rami Mustapha
- Univ. Lille, Inserm, CHU Lille, U1189 - ONCO-THAI - Assisted Laser Therapy and Immunotherapy for Oncology, Lille, France
- Department of Cancer Studies and Pharmaceutical Sciences New Hunt’s House, School of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Belinda Duchêne
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER – Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, France
| | - Abhishek Kumar
- Univ. Lille, Inserm, CHU Lille, U1189 - ONCO-THAI - Assisted Laser Therapy and Immunotherapy for Oncology, Lille, France
| | - Elisabeth Werkmeister
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UMS 2014 -PLBS, Lille, France
| | - Emmanuelle Leteurtre
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER – Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, France
| | - Olivier Moralès
- Univ. Lille, Inserm, CHU Lille, U1189 - ONCO-THAI - Assisted Laser Therapy and Immunotherapy for Oncology, Lille, France
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER – Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, France
| | - Nicolas Jonckheere
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER – Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, France
| | - Isabelle Van Seuningen
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER – Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, France
| | - Nadira Delhem
- Univ. Lille, Inserm, CHU Lille, U1189 - ONCO-THAI - Assisted Laser Therapy and Immunotherapy for Oncology, Lille, France
| |
Collapse
|
6
|
Jee W, Ko HM, Park DI, Park YR, Park SM, Kim H, Na YC, Jung JH, Jang HJ. Momordicae Semen inhibits migration and induces apoptotic cell death by regulating c-Myc and CNOT2 in human pancreatic cancer cells. Sci Rep 2023; 13:12800. [PMID: 37550432 PMCID: PMC10406802 DOI: 10.1038/s41598-023-39840-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: 11/30/2022] [Accepted: 07/31/2023] [Indexed: 08/09/2023] Open
Abstract
Pancreatic cancer(PC) is less common than other cancers; however, it has a poor prognosis. Therefore, studying novel target signaling and anticancer agents is necessary. Momordicae Semen (MS), the seed of Momordica sochinensis Spreng, mainly found in South-East Asia, including China and Bangladesh, is used to treat various diseases because of its anticancer, antioxidant, anti-inflammatory, and antibacterial properties. However, the effect of the MS extract on pancreatic cancer cells remains unknown. In this study investigated whether the MS extract exerted an anti-cancer effect by regulating c-Myc through CNOT2. Cytotoxicity and proliferation were investigated using MTT and colony formation assays. The levels of apoptotic, oncogenic, and migration-associated factors were confirmed using immunoblotting and immunofluorescence. Wound closure was analyzed using a wound healing assay. The chemical composition of the MS methanol extracts was analyzed using liquid chromatography-mass spectrometry. We confirmed that the MS extract regulated apoptotic factors and attenuated the stability of c-Myc and its sensitivity to fetal bovine serum. Furthermore, the MS extract increased apoptosis by regulating c-Myc and CNOT2 expression and enhanced the sensitivity of 5-FU in pancreatic cancer. This study showed that the MS extract is a promising new drug for PC.
Collapse
Affiliation(s)
- Wona Jee
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Korea
| | - Hyun Min Ko
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Korea
| | - Do-Il Park
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Korea
| | - Ye-Rin Park
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Korea
| | - So-Mi Park
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Korea
| | - Hyungsuk Kim
- Department of Korean Rehabilitation Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Yun-Cheol Na
- Western Seoul Center, Korea Basic Science Institute, 150 Bugahyeon-Ro, Seodaemun-Gu, Seoul, 03759, Republic of Korea
| | - Ji Hoon Jung
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Korea
| | - Hyeung-Jin Jang
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, Korea.
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Korea.
| |
Collapse
|
7
|
Chamma H, Vila IK, Taffoni C, Turtoi A, Laguette N. Activation of STING in the pancreatic tumor microenvironment: A novel therapeutic opportunity. Cancer Lett 2022; 538:215694. [PMID: 35489447 DOI: 10.1016/j.canlet.2022.215694] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/21/2022] [Accepted: 04/15/2022] [Indexed: 12/20/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a cancer of poor prognosis that presents with a dense desmoplastic stroma that contributes to therapeutic failure. PDAC patients are mostly unresponsive to immunotherapy. However, hopes to elicit response to immunotherapy have emerged with novel strategies targeting the Stimulator of Interferon Genes (STING) protein, which is a major regulator of tumor-associated inflammation. Combination of STING agonists with conventional immunotherapy approaches has proven to potentiate therapeutic benefits in several cancers. However, recent data underscore that the output of STING activation varies depending on the cellular and tissue context. This suggests that tumor heterogeneity, and in particular the heterogeneity of the tumor microenvironment (TME), is a key factor determining whether STING activation would bear benefits for patients. In this review, we discuss the potential benefits of STING activation in PDAC. To this aim, we describe the major components of the PDAC TME, and the expected consequences of STING activation.
Collapse
Affiliation(s)
- Hanane Chamma
- Institut de Génétique Humaine, CNRS, Université de Montpellier, Molecular Basis of Inflammation Laboratory, Montpellier, France
| | - Isabelle K Vila
- Institut de Génétique Humaine, CNRS, Université de Montpellier, Molecular Basis of Inflammation Laboratory, Montpellier, France
| | - Clara Taffoni
- Institut de Génétique Humaine, CNRS, Université de Montpellier, Molecular Basis of Inflammation Laboratory, Montpellier, France
| | - Andrei Turtoi
- Tumor Microenvironment Laboratory, Institut de Recherche en Cancérologie de Montpellier, Université de Montpellier, INSERM U1194, 34000, Montpellier, France.
| | - Nadine Laguette
- Institut de Génétique Humaine, CNRS, Université de Montpellier, Molecular Basis of Inflammation Laboratory, Montpellier, France.
| |
Collapse
|
8
|
Wang K, Herr I. Machine-Learning-Based Bibliometric Analysis of Pancreatic Cancer Research Over the Past 25 Years. Front Oncol 2022; 12:832385. [PMID: 35419289 PMCID: PMC8995465 DOI: 10.3389/fonc.2022.832385] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 03/03/2022] [Indexed: 12/21/2022] Open
Abstract
Machine learning and semantic analysis are computer-based methods to evaluate complex relationships and predict future perspectives. We used these technologies to define recent, current and future topics in pancreatic cancer research. Publications indexed under the Medical Subject Headings (MeSH) term 'Pancreatic Neoplasms' from January 1996 to October 2021 were downloaded from PubMed. Using the statistical computing language R and the interpreted, high-level, general-purpose programming language Python, we extracted publication dates, geographic information, and abstracts from each publication's metadata for bibliometric analyses. The generative statistical algorithm "latent Dirichlet allocation" (LDA) was applied to identify specific research topics and trends. The unsupervised "Louvain algorithm" was used to establish a network to identify relationships between single topics. A total of 60,296 publications were identified and analyzed. The publications were derived from 133 countries, mostly from the Northern Hemisphere. For the term "pancreatic cancer research", 12,058 MeSH terms appeared 1,395,060 times. Among them, we identified the four main topics "Clinical Manifestation and Diagnosis", "Review and Management", "Treatment Studies", and "Basic Research". The number of publications has increased rapidly during the past 25 years. Based on the number of publications, the algorithm predicted that "Immunotherapy", Prognostic research", "Protein expression", "Case reports", "Gemcitabine and mechanism", "Clinical study of gemcitabine", "Operation and postoperation", "Chemotherapy and resection", and "Review and management" as current research topics. To our knowledge, this is the first study on this subject of pancreatic cancer research, which has become possible due to the improvement of algorithms and hardware.
Collapse
Affiliation(s)
- Kangtao Wang
- Molecular OncoSurgery, Section Surgical Research, Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Ingrid Herr
- Molecular OncoSurgery, Section Surgical Research, Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
9
|
Qorri B, Mokhtari RB, Harless WW, Szewczuk MR. Next Generation of Cancer Drug Repurposing: Therapeutic Combination of Aspirin and Oseltamivir Phosphate Potentiates Gemcitabine to Disable Key Survival Pathways Critical for Pancreatic Cancer Progression. Cancers (Basel) 2022; 14:1374. [PMID: 35326525 PMCID: PMC8946854 DOI: 10.3390/cancers14061374] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/26/2022] [Accepted: 03/02/2022] [Indexed: 12/12/2022] Open
Abstract
Resistance to chemotherapeutics and high metastatic rates contribute to the abysmal survival rate in patients with pancreatic cancer. An alternate approach for treating human pancreatic cancer involves repurposing the anti-inflammatory drug, aspirin (ASA), with oseltamivir phosphate (OP) in combination with the standard chemotherapeutic agent, gemcitabine (GEM). The question is whether treatment with ASA and OP can sensitize cancer cells to the cytotoxicity induced by GEM and limit the development of chemoresistance. To assess the key survival pathways critical for pancreatic cancer progression, we used the AlamarBlue cytotoxicity assay to determine the cell viability and combination index for the drug combinations, flow cytometric analysis of annexin V apoptosis assay to detect apoptotic and necrotic cells, fluorometric QCM™ chemotaxis migration assay to assess cellular migration, fluorometric extracellular matrix (ECM) cell adhesion array kit to assess the expression of the ECM proteins, scratch wound assay using the 96-well WoundMaker™, and the methylcellulose clonogenic assay to assess clonogenic potential. The combination of ASA and OP with GEM significantly upended MiaPaCa-2 and PANC-1 pancreatic cancer cell viability, clonogenic potential, expression of critical extracellular matrix proteins, migration, and promoted apoptosis. ASA in combination with OP significantly improves the effectiveness of GEM in the treatment of pancreatic cancer and disables key survival pathways critical to disease progression.
Collapse
Affiliation(s)
- Bessi Qorri
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (B.Q.); (R.B.M.)
| | - Reza Bayat Mokhtari
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (B.Q.); (R.B.M.)
| | | | - Myron R. Szewczuk
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (B.Q.); (R.B.M.)
| |
Collapse
|
10
|
Zhang Z, Yang A, Chaurasiya S, Park AK, Kim SI, Lu J, Olafsen T, Warner SG, Fong Y, Woo Y. PET imaging and treatment of pancreatic cancer peritoneal carcinomatosis after subcutaneous intratumoral administration of a novel oncolytic virus, CF33-hNIS-antiPDL1. Mol Ther Oncolytics 2022; 24:331-339. [PMID: 35118191 PMCID: PMC8784298 DOI: 10.1016/j.omto.2021.12.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 12/28/2021] [Indexed: 11/10/2022] Open
Abstract
Peritoneal carcinomatosis of gastrointestinal malignancies remains fatal. CF33-hNIS-antiPDL1, a chimeric orthopoxvirus expressing the human sodium iodide symporter (hNIS) and anti-human programmed death-ligand 1 antibody, has demonstrated robust preclinical activity against pancreatic adenocarcinoma (PDAC). We investigated the ability of CF33-hNIS-antiPDL1 to infect, help detect, and kill peritoneal tumors following intratumoral (i.t.) injection of subcutaneous (s.c.) tumors in vivo. Human PDAC AsPC-1-ffluc cells were inoculated in both the s.c. space and the peritoneal cavity of athymic mice. After successful tumor engraftment, s.c. tumors were injected with CF33-hNIS-antiPDL1 or PBS. We assessed the ability of CF33-hNIS-antiPDL1 to infect, replicate in, and allow the imaging of tumors at both sites (immunohistochemistry [IHC] and 124I-based positron emission tomography/computed tomography [PET/CT] imaging), tumor burden (bioluminescence imaging), and animal survival. IHC staining for hNIS confirmed expression in s.c. and peritoneal tumors following virus treatment. Compared to the controls, CF33-hNIS-antiPDL1-treated mice showed significantly decreased s.c. and peritoneal tumor burden and improved survival (p < 0.05). Notably, 2 of 8 mice showed complete regression of disease. PET/CT avidity for 124I uptake in s.c. and peritoneal tumors was visible starting at day 7 following the first i.t. dose of CF33-hNIS-antiPDL1. We show that CF33-hNIS-antiPDL1 can help detect and kill both s.c. and peritoneal tumors following s.c. i.t. treatment.
Collapse
|
11
|
Khan NN, Lewin T, Hatton A, Pilgrim C, Ioannou L, Te Marvelde L, Zalcberg J, Evans S. Systematic review of the predictors of health service use in pancreatic cancer. Am J Cancer Res 2022; 12:622-650. [PMID: 35261792 PMCID: PMC8900007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/21/2022] [Indexed: 06/14/2023] Open
Abstract
INTRODUCTION Pancreatic cancer (PC) has a dismal prognosis, with identified disparities in survival outcomes based on demographic characteristics. These disparities may be ameliorated by equitable access to treatments and health services. This systematic review identifies patient and service-level characteristics associated with PC health service utilisation (HSU). METHODS Medline, Embase, CINAHL, PsycINFO and Scopus were systematically searched between 1st January, 2010 and 17 May, 2021 for population-based, PC studies which conducted univariable and/or multivariable regression analyses to identify patient and/or service-level characteristics associated with use of a treatment or health service. Direction of effect sizes were reported in an aggregate manner. RESULTS Sixty-two eligible studies were identified. Most (48/62) explored the predictors of surgery (n=25) and chemotherapy (n=23), and in populations predominantly based in the United States of America (n=50). Decreased HSU was observed among people belonging to older age groups, non-Caucasian ethnicities, lower socioeconomic status (SES) and lower education status. Non-metropolitan location of residence predicted decreased use of certain treatments, and was associated with reduced hospitalisations. People with comorbidities were less likely to use treatments and services, including specialist consultations and palliative care but were more likely to be hospitalised. A more recent year of diagnosis/year of death was generally associated with increased HSU. Academically affiliated and high-volume centres predicted increased treatment use and hospital readmissions. CONCLUSION Findings of this review may assist identification of vulnerable patient groups experiencing disparities in accessing and using treatments and therapies.
Collapse
Affiliation(s)
- Nadia N Khan
- Public Health and Preventive Medicine, Monash University Melbourne, Victoria, Australia
| | - Tennille Lewin
- Public Health and Preventive Medicine, Monash University Melbourne, Victoria, Australia
| | - Amy Hatton
- Public Health and Preventive Medicine, Monash University Melbourne, Victoria, Australia
| | - Charles Pilgrim
- Public Health and Preventive Medicine, Monash University Melbourne, Victoria, Australia
| | - Liane Ioannou
- Public Health and Preventive Medicine, Monash University Melbourne, Victoria, Australia
| | - Luc Te Marvelde
- Public Health and Preventive Medicine, Monash University Melbourne, Victoria, Australia
| | - John Zalcberg
- Public Health and Preventive Medicine, Monash University Melbourne, Victoria, Australia
| | - Sue Evans
- Public Health and Preventive Medicine, Monash University Melbourne, Victoria, Australia
| |
Collapse
|