1
|
Chen Z, Wang J, Kankala RK, Jiang M, Long L, Li W, Zou L, Chen A, Liu Y. Decellularized extracellular matrix-based disease models for drug screening. Mater Today Bio 2024; 29:101280. [PMID: 39399243 PMCID: PMC11470555 DOI: 10.1016/j.mtbio.2024.101280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/12/2024] [Accepted: 09/26/2024] [Indexed: 10/15/2024] Open
Abstract
In vitro drug screening endeavors to replicate cellular states closely resembling those encountered in vivo, thereby maximizing the fidelity of drug effects and responses within the body. Decellularized extracellular matrix (dECM)-based materials offer a more authentic milieu for crafting disease models, faithfully emulating the extracellular components and structural complexities encountered by cells in vivo. This review discusses recent advancements in leveraging dECM-based materials as biomaterials for crafting cell models tailored for drug screening. Initially, we delineate the biological functionalities of diverse ECM components, shedding light on their potential influences on disease model construction. Further, we elucidate the decellularization techniques and methodologies for fabricating cell models utilizing dECM substrates. Then, the article delves into the research strides made in employing dECM-based models for drug screening across a spectrum of ailments, including tumors, as well as heart, liver, lung, and bone diseases. Finally, the review summarizes the bottlenecks, hurdles, and promising research trajectories associated with the dECM materials for drug screening, alongside their prospective applications in personalized medicine. Together, by encapsulating the contemporary research landscape surrounding dECM materials in cell model construction and drug screening, this review underscores the vast potential of dECM materials in drug assessment and personalized therapy.
Collapse
Affiliation(s)
- Zhoujiang Chen
- Institute for Advanced Study, Chengdu University, Chengdu, 610106, Sichuan, PR China
| | - Ji Wang
- Affiliated Hospital & Clinical Medical College of Chengdu University, Chengdu University, Chengdu, 610106, Sichuan, PR China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, PR China
| | - Mingli Jiang
- School of Pharmacy, Zunyi Medical University, Zunyi, 563099, Guizhou, PR China
| | - Lianlin Long
- School of Pharmacy, Zunyi Medical University, Zunyi, 563099, Guizhou, PR China
| | - Wei Li
- Affiliated Hospital & Clinical Medical College of Chengdu University, Chengdu University, Chengdu, 610106, Sichuan, PR China
| | - Liang Zou
- Institute for Advanced Study, Chengdu University, Chengdu, 610106, Sichuan, PR China
| | - Aizheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, PR China
| | - Ya Liu
- Affiliated Hospital & Clinical Medical College of Chengdu University, Chengdu University, Chengdu, 610106, Sichuan, PR China
| |
Collapse
|
2
|
Xiao Y, Pan T, Da W, Liu Y, Chen S, Chen D, Liu K, Zheng Y, Xie D, Gao Y, Xu H, Sun Y, Tan W. Aptamer-drug conjugates-loaded bacteria for pancreatic cancer synergistic therapy. Signal Transduct Target Ther 2024; 9:272. [PMID: 39397032 PMCID: PMC11471780 DOI: 10.1038/s41392-024-01973-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/30/2024] [Accepted: 09/11/2024] [Indexed: 10/15/2024] Open
Abstract
Pancreatic cancer is one of the most malignant tumors with the highest mortality rates, and it currently lacks effective drugs. Aptamer-drug conjugates (ApDC), as a form of nucleic acid drug, show great potential in cancer therapy. However, the instability of nucleic acid-based drugs in vivo and the avascularity of pancreatic cancer with dense stroma have limited their application. Fortunately, VNP20009, a genetically modified strain of Salmonella typhimurium, which has a preference for anaerobic environments, but is toxic and lacks specificity, can potentially serve as a delivery vehicle for ApDC. Here, we propose a synergistic therapy approach that combines the penetrative capability of bacteria with the targeting and toxic effects of ApDC by conjugating ApDC to VNP20009 through straightforward, one-step click chemistry. With this strategy, bacteria specifically target pancreatic cancer through anaerobic chemotaxis and subsequently adhere to tumor cells driven by the aptamer's specific binding. Results indicate that this method prolongs the serum stability of ApDC up to 48 h and resulted in increased drug concentration at tumor sites compared to the free drugs group. Moreover, the aptamer's targeted binding to cancer cells tripled bacterial colonization at the tumor site, leading to increased death of tumor cells and T cell infiltration. Notably, by integrating chemotherapy and immunotherapy, the effectiveness of the treatment is significantly enhanced, showing consistent results across various animal models. Overall, this strategy takes advantage of bacteria and ApDC and thus presents an effective synergistic strategy for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Yu Xiao
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Pan
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wuren Da
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuanding Liu
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shuangya Chen
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Daiquan Chen
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Keying Liu
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yihan Zheng
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Daolong Xie
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuan Gao
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haiyan Xu
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Yang Sun
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China.
| |
Collapse
|
3
|
Miller CS, Lecavalier-Barsoum M, Ma K, Santos Dutra M, Kaitoukov Y, Bahoric B, Tomic N, Dinelle F, Enger S, Batist G, Yang S, Laporta D, Kavan P, Sahai A, Roberge D, Donath D. Feasibility and safety of endoscopic ultrasound-guided diffusing alpha emitter radiation therapy for advanced pancreatic cancer: Preliminary data. Endosc Int Open 2024; 12:E1085-E1091. [PMID: 39398444 PMCID: PMC11469937 DOI: 10.1055/a-2379-1591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/02/2024] [Indexed: 10/15/2024] Open
Abstract
Background and study aims Pancreatic cancer is a devastating disease with limited locoregional treatment options. Diffusing alpha-emitter radiation therapy (Alpha DaRT), a novel cancer treatment using alpha-particle interstitial radiotherapy, may help address this challenge. The aim of this study was to evaluate the feasibility and safety of endoscopic ultrasound (EUS)-guided Alpha DaRT for advanced pancreatic cancer. Patients and methods Patients with inoperable locally advanced or metastatic pancreatic adenocarcinoma were treated with EUS-guided Alpha DaRT insertion. The Alpha DaRT sources were delivered into pancreatic tumors using a standard EUS needle with a novel proprietary applicator. Adverse events (AEs) were assessed based on the Common Terminology Criteria for Adverse Events version 5.0. Tumor response was evaluated by imaging 4 to 6 weeks post treatment. Results The first five patients were treated between March and September 2023. The procedure was technically successful in all cases, with Alpha DaRT sources inserted into the target tumor. Estimated gross tumor volume coverage ranged from 8% to 44%. Fourteen AEs were reported among three patients. Four were serious AEs, none of which was associated with the treatment, but rather, with disease progression or medical assistance in dying. Only two AEs (mild) were deemed possibly related to the study device. At the 35-day visit, two patients had progressive disease and three had stable disease, with one of the latter showing partial response 2 months post procedure. Conclusions Preliminary results from this first-in-human trial indicate that EUS-guided Alpha DaRT treatment for unresectable pancreatic cancer is feasible and safe, with no device-associated serious AEs. Further investigation of this promising novel modality is underway.
Collapse
Affiliation(s)
- Corey S Miller
- Gastroenterology, Jewish General Hospital, Montreal, Canada
- Medicine, McGill University Faculty of Medicine, Montreal, Canada
- McGill Centre for Translational Research in Cancer, Lady Davis Institute for Medical Research, Montreal, Canada
| | - Magali Lecavalier-Barsoum
- Medicine, McGill University Faculty of Medicine, Montreal, Canada
- McGill Centre for Translational Research in Cancer, Lady Davis Institute for Medical Research, Montreal, Canada
- Radiation Oncology, Jewish General Hospital, Montreal, Canada
| | - Kim Ma
- Medicine, McGill University Faculty of Medicine, Montreal, Canada
- Oncology, Jewish General Hospital, Montreal, Canada
| | - Miriam Santos Dutra
- McGill Centre for Translational Research in Cancer, Lady Davis Institute for Medical Research, Montreal, Canada
| | - Youri Kaitoukov
- Medicine, McGill University Faculty of Medicine, Montreal, Canada
- Radiology, Jewish General Hospital, Montreal, Canada
| | - Boris Bahoric
- McGill Centre for Translational Research in Cancer, Lady Davis Institute for Medical Research, Montreal, Canada
- Radiation Oncology, Jewish General Hospital, Montreal, Canada
| | - Nada Tomic
- Medical Physics Unit, McGill University, Montreal, Canada
| | - Francine Dinelle
- Quality and Transformation Unit, Jewish General Hospital, Montreal, Canada
| | - Shirin Enger
- McGill Centre for Translational Research in Cancer, Lady Davis Institute for Medical Research, Montreal, Canada
- Oncology, Jewish General Hospital, Montreal, Canada
- Medical Physics Unit, McGill University, Montreal, Canada
| | - Gerald Batist
- McGill Centre for Translational Research in Cancer, Lady Davis Institute for Medical Research, Montreal, Canada
- Oncology, Jewish General Hospital, Montreal, Canada
| | - Stephen Yang
- Medicine, McGill University Faculty of Medicine, Montreal, Canada
- Anesthesia, Jewish General Hospital, Montreal, Canada
| | - Donald Laporta
- Medicine, McGill University Faculty of Medicine, Montreal, Canada
- Adult Critical Care and Respirology, Jewish General Hospital, Montreal, Canada
| | - Petr Kavan
- McGill Centre for Translational Research in Cancer, Lady Davis Institute for Medical Research, Montreal, Canada
- Oncology, Jewish General Hospital, Montreal, Canada
| | - Anand Sahai
- Gastroenterology, Centre Hospitalier de l’Université de Montréal, Montreal, Canada
| | - David Roberge
- Radiation Oncology, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
| | - David Donath
- Radiation Oncology, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
| |
Collapse
|
4
|
Petrelli F, Rosenfeld R, Ghidini A, Celotti A, Dottorini L, Viti M, Baiocchi G, Garrone O, Tomasello G, Ghidini M. Comparative Efficacy of 21 Treatment Strategies for Resectable Pancreatic Cancer: A Network Meta-Analysis. Cancers (Basel) 2024; 16:3203. [PMID: 39335177 PMCID: PMC11429569 DOI: 10.3390/cancers16183203] [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: 08/23/2024] [Revised: 09/14/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
The primary treatment for operable pancreatic cancer (PC) involves surgery followed by adjuvant therapy. Nevertheless, perioperative or neoadjuvant chemotherapy (CT) may be used to mitigate the likelihood of recurrence and mortality. This network meta-analysis (NMA) assesses the comparative efficacy of various treatment approaches for resectable PC. A thorough search was carried out on January 31, 2023, encompassing PubMed/MEDLINE, Cochrane Library, and Embase databases. We incorporated randomized clinical trials (RCTs) that compared surgical interventions with or without (neo)adjuvant or perioperative therapies for operable PC. We conducted a fixed-effects Bayesian NMA. We presented the effect sizes in terms of hazard ratios (HRs) for overall survival (OS) along with 95% credible intervals (95% CrIs). The treatment was deemed statistically superior when the 95% credible interval (CrI) did not encompass a null value (hazard ratio < 1). Treatment rankings were established based on the surface under the cumulative ranking curve (SUCRA). A total of 24 studies were incorporated, comparing 21 treatments with surgery in isolation. Eleven treatments showed superior efficacy compared to surgery alone, with HRs ranging from 0.38 for perioperative treatments to 0.73 for adjuvant 5-fluorouracil. After the exclusion of studies conducted in Asia, it was found that the perioperative regimen of gemcitabine combined with nab-paclitaxel was the most effective regimen (SUCRA, p = 0.99). The findings endorse the utilization of perioperative CT, especially multi-agent CT, as the favored intervention for operable PC in Western nations.
Collapse
Affiliation(s)
- Fausto Petrelli
- Oncology Unit, Oncology Department, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047 Treviglio, Italy;
| | - Roberto Rosenfeld
- Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (R.R.); (O.G.); (M.G.)
| | | | - Andrea Celotti
- Surgery Unit, ASST Cremona, 26100 Cremona, Italy; (A.C.); (G.B.)
| | - Lorenzo Dottorini
- Oncology Unit, Oncology Department, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047 Treviglio, Italy;
| | - Matteo Viti
- Surgery Unit, ASST Bergamo Ovest, 24047 Treviglio, Italy;
| | | | - Ornella Garrone
- Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (R.R.); (O.G.); (M.G.)
| | | | - Michele Ghidini
- Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (R.R.); (O.G.); (M.G.)
| |
Collapse
|
5
|
Minaei E, Ranson M, Aghmesheh M, Sluyter R, Vine KL. Enhancing pancreatic cancer immunotherapy: Leveraging localized delivery strategies through the use of implantable devices and scaffolds. J Control Release 2024; 373:145-160. [PMID: 38996923 DOI: 10.1016/j.jconrel.2024.07.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/03/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
Pancreatic cancer (PC) remains the predominant type of upper gastrointestinal tract cancer, associated with heightened morbidity and a survival rate below 12%. While immunotherapy has brought about transformative changes in the standards of care for most solid tumors, its application in PC is hindered by the ''cold tumor'' microenvironment, marked by the presence of immunosuppressive cells. Modest response rates in PC are attributed, in part to, the fibrotic stroma that obstructs the delivery of systemic immunotherapy. Furthermore, the occurrence of immune-related adverse events (iRAEs) often necessitates the use of sub-therapeutic doses or treatment discontinuation. In the pursuit of innovative approaches to enhance the effectiveness of immunotherapy for PC, implantable drug delivery devices and scaffolds emerge as promising strategies. These technologies offer the potential for sustained drug delivery directly to the tumor site, overcoming stromal barriers, immunosuppression, T cell exclusion, immunotherapy resistance, optimizing drug dosage, and mitigating systemic toxicity. This review offers a comprehensive exploration of pancreatic ductal adenocarcinoma (PDAC), the most common and aggressive form of PC, accompanied by a critical analysis of the challenges the microenvironment presents to the development of successful combinational immunotherapy approaches. Despite efforts, these approaches have thus far fallen short in enhancing treatment outcomes for PDAC. The review will subsequently delve into the imperative need for refining delivery strategies, providing an examination of past and ongoing studies in the field of localized immunotherapy for PDAC. Addressing these issues will lay the groundwork for the development of effective new therapies, thereby enhancing treatment response, patient survival, and overall quality of life for individuals diagnosed with PDAC.
Collapse
Affiliation(s)
- E Minaei
- School of Chemistry and Molecular Bioscience, Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia.
| | - M Ranson
- School of Chemistry and Molecular Bioscience, Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - M Aghmesheh
- Nelune Comprehensive Cancer Centre, Bright Building, Prince of Wales Hospital, Sydney, NSW, Australia; Faculty of Medicine and Health, UNSW, Sydney, NSW, Australia
| | - R Sluyter
- School of Chemistry and Molecular Bioscience, Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - K L Vine
- School of Chemistry and Molecular Bioscience, Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia.
| |
Collapse
|
6
|
Pecoraro C, Carbone D, Scianò F, Terrana F, Xu G, Bergonzini C, Roeten MSF, Cascioferro S, Cirrincione G, Diana P, Giovannetti E, Parrino B. Exploring the therapeutic potential of a novel series of imidazothiadiazoles targeting focal adhesion kinase (FAK) for pancreatic cancer treatment: synthesis, mechanistic insights and promising antitumor and safety profile. J Drug Target 2024:1-17. [PMID: 39067009 DOI: 10.1080/1061186x.2024.2385557] [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: 04/22/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Focal Adhesion Kinase (FAK) is a non-receptor protein tyrosine kinase that plays a crucial role in various oncogenic processes related to cell adhesion, migration, proliferation, and survival. The strategic targeting of FAK represents a burgeoning approach to address resistant tumours, such as pancreatic ductal adenocarcinoma (PDAC). Herein, we report a new series of twenty imidazo[2,1-b][1, 3, 4]thiadiazole derivatives assayed for their antiproliferative activity against the National Cancer Institute (NCI-60) panel and a wide panel of PDAC models. Lead compound 10l exhibited effective antiproliferative activity against immortalised (SUIT-2, CAPAN-1, PANC-1, PATU-T, BxPC-3), primary (PDAC-3) and gemcitabine-resistant clone (PANC-1-GR) PDAC cells, eliciting IC50 values in the low micromolar range (1.04-3.44 µM), associated with a significant reduction in cell-migration and spheroid shrinkage in vitro. High-throughput kinase arrays revealed a significant inhibition of the FAK signalling network, associated to induction of cell cycle arrest in G2/M phase, suppression of tumour cell invasion and apoptosis induction. The high selectivity index/toxicity prompted studies using PDAC mouse xenografts, demonstrating significant inhibition of tumour growth and safety. In conclusion, compound 10l displayed antitumor activity and safety in both in vitro and in vivo models, emerging as a highly promising lead for the development of FAK inhibitors in PDAC.
Collapse
Affiliation(s)
- Camilla Pecoraro
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Palermo, Italy
| | - Daniela Carbone
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Palermo, Italy
| | - Fabio Scianò
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University, Amsterdam, The Netherlands
- Lumobiotics, Karlsruhe, Germany
| | - Francesca Terrana
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Palermo, Italy
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University, Amsterdam, The Netherlands
| | - Geng Xu
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University, Amsterdam, The Netherlands
| | - Cecilia Bergonzini
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University, Amsterdam, The Netherlands
- Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Margot S F Roeten
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, The Netherlands
| | - Stella Cascioferro
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Palermo, Italy
| | - Girolamo Cirrincione
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Palermo, Italy
| | - Patrizia Diana
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Palermo, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University, Amsterdam, The Netherlands
- Cancer Pharmacology Laboratory, Fondazione Pisana per la Scienza, Pisa, Italy
| | - Barbara Parrino
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Palermo, Italy
| |
Collapse
|
7
|
Arayici ME, İnal A, Basbinar Y, Olgun N. Evaluation of the diagnostic and prognostic clinical values of circulating tumor DNA and cell-free DNA in pancreatic malignancies: a comprehensive meta-analysis. Front Oncol 2024; 14:1382369. [PMID: 38983931 PMCID: PMC11231086 DOI: 10.3389/fonc.2024.1382369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/10/2024] [Indexed: 07/11/2024] Open
Abstract
Background The diagnostic and prognostic clinical value of circulating tumor DNA (ctDNA) and cell-free DNA (cfDNA) in pancreatic malignancies are unclear. Herein, we aimed to perform a meta-analysis to evaluate ctDNA and cfDNA as potential diagnostic and prognostic biomarkers. Methods PRISMA reporting guidelines were followed closely for conducting the current meta-analysis. The PubMed/Medline, Scopus, and Web of Science (WoS) databases were scanned in detail to identify eligible papers for the study. A quality assessment was performed in accordance with the REMARK criteria. The risk ratios (RRs) of the diagnostic accuracy of ctDNA compared to that of carbohydrate antigen 19.9 (CA 19.9) in all disease stages and the hazard ratios (HRs) of the prognostic role of ctDNA in overall survival (OS) were calculated with 95% confidence intervals (CIs). Results A total of 18 papers were evaluated to assess the diagnostic accuracy and prognostic value of biomarkers related to pancreatic malignancies. The pooled analysis indicated that CA19.9 provides greater diagnostic accuracy across all disease stages than ctDNA or cfDNA (RR = 0.64, 95% CI: 0.50-0.82, p < 0.001). Additionally, in a secondary analysis focusing on prognosis, patients who were ctDNA-positive were found to have significantly worse OS (HR = 2.00, 95% CI: 1.51-2.66, p < 0.001). Conclusion The findings of this meta-analysis demonstrated that CA19-9 still has greater diagnostic accuracy across all disease stages than KRAS mutations in ctDNA or cfDNA. Nonetheless, the presence of detectable levels of ctDNA was associated with worse patient outcomes regarding OS. There is a growing need for further research on this topic. Systematic review registration https://doi.org/10.37766/inplasy2023.12.0092, identifier INPLASY2023120092.
Collapse
Affiliation(s)
- Mehmet Emin Arayici
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Dokuz Eylül University, İzmir, Türkiye
- Department of Public Health, Faculty of Medicine, Dokuz Eylül University, İzmir, Türkiye
| | - Abdullah İnal
- Department of General Surgery, Faculty of Medicine, İzmir Democracy University, İzmir, Türkiye
| | - Yasemin Basbinar
- Department of Translational Oncology, Institute of Oncology, Dokuz Eylül University, İzmir, Türkiye
| | - Nur Olgun
- Department of Clinical Oncology, Institute of Oncology, Dokuz Eylül University, İzmir, Türkiye
| |
Collapse
|
8
|
Han X, Cai C, Deng W, Shi Y, Li L, Wang C, Zhang J, Rong M, Liu J, Fang B, He H, Liu X, Deng C, He X, Cao X. Landscape of human organoids: Ideal model in clinics and research. Innovation (N Y) 2024; 5:100620. [PMID: 38706954 PMCID: PMC11066475 DOI: 10.1016/j.xinn.2024.100620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/29/2024] [Indexed: 05/07/2024] Open
Abstract
In the last decade, organoid research has entered a golden era, signifying a pivotal shift in the biomedical landscape. The year 2023 marked a milestone with the publication of thousands of papers in this arena, reflecting exponential growth. However, amid this burgeoning expansion, a comprehensive and accurate overview of the field has been conspicuously absent. Our review is intended to bridge this gap, providing a panoramic view of the rapidly evolving organoid landscape. We meticulously analyze the organoid field from eight distinctive vantage points, harnessing our rich experience in academic research, industrial application, and clinical practice. We present a deep exploration of the advances in organoid technology, underpinned by our long-standing involvement in this arena. Our narrative traverses the historical genesis of organoids and their transformative impact across various biomedical sectors, including oncology, toxicology, and drug development. We delve into the synergy between organoids and avant-garde technologies such as synthetic biology and single-cell omics and discuss their pivotal role in tailoring personalized medicine, enhancing high-throughput drug screening, and constructing physiologically pertinent disease models. Our comprehensive analysis and reflective discourse provide a deep dive into the existing landscape and emerging trends in organoid technology. We spotlight technological innovations, methodological evolution, and the broadening spectrum of applications, emphasizing the revolutionary influence of organoids in personalized medicine, oncology, drug discovery, and other fields. Looking ahead, we cautiously anticipate future developments in the field of organoid research, especially its potential implications for personalized patient care, new avenues of drug discovery, and clinical research. We trust that our comprehensive review will be an asset for researchers, clinicians, and patients with keen interest in personalized medical strategies. We offer a broad view of the present and prospective capabilities of organoid technology, encompassing a wide range of current and future applications. In summary, in this review we attempt a comprehensive exploration of the organoid field. We offer reflections, summaries, and projections that might be useful for current researchers and clinicians, and we hope to contribute to shaping the evolving trajectory of this dynamic and rapidly advancing field.
Collapse
Affiliation(s)
- Xinxin Han
- Organ Regeneration X Lab, Lisheng East China Institute of Biotechnology, Peking University, Jiangsu 226200, China
- Shanghai Lisheng Biotech, Shanghai 200092, China
| | - Chunhui Cai
- Shanghai Lisheng Biotech, Shanghai 200092, China
| | - Wei Deng
- LongHua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wanping South Road, Xuhui District, Shanghai 200032, China
- Department of Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Yanghua Shi
- Shanghai Lisheng Biotech, Shanghai 200092, China
| | - Lanyang Li
- Shanghai Lisheng Biotech, Shanghai 200092, China
| | - Chen Wang
- Shanghai Lisheng Biotech, Shanghai 200092, China
| | - Jian Zhang
- Shanghai Lisheng Biotech, Shanghai 200092, China
| | - Mingjie Rong
- Shanghai Lisheng Biotech, Shanghai 200092, China
| | - Jiping Liu
- Shanghai Lisheng Biotech, Shanghai 200092, China
| | - Bangjiang Fang
- LongHua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wanping South Road, Xuhui District, Shanghai 200032, China
| | - Hua He
- Department of Neurosurgery, Third Affiliated Hospital, Naval Medical University, Shanghai 200438, China
| | - Xiling Liu
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Ministry of Justice, Shanghai 200063, China
| | - Chuxia Deng
- Cancer Center, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
- Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR 999078, China
| | - Xiao He
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Cao
- Zhongshan Hospital Institute of Clinical Science, Fudan University Shanghai Medical College, Shanghai 200032, China
| |
Collapse
|
9
|
Wang H, Wang Y, Zhang D, Li P. Circulating nucleosomes as potential biomarkers for cancer diagnosis and treatment monitoring. Int J Biol Macromol 2024; 262:130005. [PMID: 38331061 DOI: 10.1016/j.ijbiomac.2024.130005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/03/2024] [Accepted: 02/04/2024] [Indexed: 02/10/2024]
Abstract
Nucleosomes play a crucial role in regulating gene expression through their composition and post-translational modifications. When cells die, intracellular endonucleases are activated and cleave chromatin into oligo- and mono-nucleosomes, which are then released into the body fluids. Studies have shown that the levels of nucleosomes are increased in serum and plasma in various cancer types, suggesting that analysis of circulating nucleosomes can provide an initial assessment of carcinogenesis. However, it should be noted that elevated serum nucleosome levels may not accurately diagnose certain tumor types, as increased cell death may occur in different pathological conditions. Nevertheless, detection of circulating nucleosomes and their histone modifications, along with specific tumor markers, can help diagnose certain types of cancer. Furthermore, monitoring changes in circulating nucleosome levels during chemotherapy or radiotherapy in patients with malignancies can provide valuable insights into clinical outcomes and therapeutic efficacy. The utilization of circulating nucleosomes as biomarkers is an exciting and emerging area of research, with the potential for early detection of various diseases and monitoring of treatment response. Integrating nucleosome-based biomarkers with existing ones may improve the specificity and sensitivity of current assays, offering the possibility of personalized precision medical treatment for patients.
Collapse
Affiliation(s)
- Huawei Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 1 Ningde Road, Qingdao 266073, China.
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 1 Ningde Road, Qingdao 266073, China.
| | - Dejiu Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 1 Ningde Road, Qingdao 266073, China.
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 1 Ningde Road, Qingdao 266073, China.
| |
Collapse
|
10
|
Shaha S, Rodrigues D, Mitragotri S. Locoregional drug delivery for cancer therapy: Preclinical progress and clinical translation. J Control Release 2024; 367:737-767. [PMID: 38325716 DOI: 10.1016/j.jconrel.2024.01.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/09/2024]
Abstract
Systemic drug delivery is the current clinically preferred route for cancer therapy. However, challenges associated with tumor localization and off-tumor toxic effects limit the clinical effectiveness of this route. Locoregional drug delivery is an emerging viable alternative to systemic therapies. With the improvement in real-time imaging technologies and tools for direct access to tumor lesions, the clinical applicability of locoregional drug delivery is becoming more prominent. Theoretically, locoregional treatments can bypass challenges faced by systemic drug delivery. Preclinically, locoregional delivery of drugs has demonstrated enhanced therapeutic efficacy with limited off-target effects while still yielding an abscopal effect. Clinically, an array of locoregional strategies is under investigation for the delivery of drugs ranging in target and size. Locoregional tumor treatment strategies can be classified into two main categories: 1) direct drug infusion via injection or implanted port and 2) extended drug elution via injected or implanted depot. The number of studies investigating locoregional drug delivery strategies for cancer treatment is rising exponentially, in both preclinical and clinical settings, with some approaches approved for clinical use. Here, we highlight key preclinical advances and the clinical relevance of such locoregional delivery strategies in the treatment of cancer. Furthermore, we critically analyze 949 clinical trials involving locoregional drug delivery and discuss emerging trends.
Collapse
Affiliation(s)
- Suyog Shaha
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Allston, MA 02134, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA
| | - Danika Rodrigues
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Allston, MA 02134, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Allston, MA 02134, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA.
| |
Collapse
|
11
|
Rahman A, Sandeep Kumar Jain R, Meghana P, Nippu BN, Manjunatha KS, Rajaput PS, Kumaraswamy HM, Satyanarayan ND. Tetrahydrobenzothiophene derivatives ameliorate Mia PaCa-2 cell progression and induces apoptosis via inhibiting EGFR2 tyrosine kinase signal. Bioorg Chem 2024; 143:106968. [PMID: 38007893 DOI: 10.1016/j.bioorg.2023.106968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/28/2023] [Accepted: 11/12/2023] [Indexed: 11/28/2023]
Abstract
A series of new thiophene analogues with acarbonitrile-basedmoiety were designed and synthesized via structural optimization. The conjugates were assessed for their in-vitro cytotoxic activity against a human pancreatic cancer cell line (Mia PaCa-2) and among them compound 5b showed IC50 value of 13.37 ± 2.37 μM. The compounds 5b (20 µM & 25 µM) and 7c (30 & 35 µM) also showed reduced clonogenicity, enhanced ROS and decreased mitochondrial membrane potential in Mia PaCa-2 cells. Treatment with these compounds also increased apoptotic population as evident with the double staining assay. Among the evaluated series, compounds 5b, 5g, 7c, and 9a attained a greater inhibitory potency than first generation's reversible EGFR inhibitor, Gefitinib. EGFR2 enzyme inhibitory studies revealed that 5b efficiently and arbitrarily suppressed the development of EGFR2 dependent cells and inhibited the enzymatic activity with an IC50 value of 0.68 µM; interestingly, the most effective molecule 5b with N-methyl piperazine substitution, has 1.29-fold greater potency than well-known EGFR inhibitor Gefitinib and increased Gefitinib's anti-growth impact with 2.04 folds greater against Mia PaCa-2. The in-vitro studies were validated with in-silico docking studies wherein compounds 5b and 7c exhibited binding energies of -8.2 and -7.4 Kcal/mol respectively. The present study reveals that tetrahydrobenzothiophene based analogues could be a promising lead for the evolution of potent chemo preventives over pancreatic cancer.
Collapse
Affiliation(s)
- Abdul Rahman
- Department of Pharmaceutical Chemistry, Kuvempu University, Post Graduate Centre, Kadur, Chikkamagaluru, Karnataka 577548, India
| | - R Sandeep Kumar Jain
- Laboratory of Experimental Medicine, Department of Biotechnology, Kuvempu University, Shankargatta, Shimoga, Karnataka 577451, India
| | - P Meghana
- Laboratory of Experimental Medicine, Department of Biotechnology, Kuvempu University, Shankargatta, Shimoga, Karnataka 577451, India
| | - B N Nippu
- Department of Pharmaceutical Chemistry, Kuvempu University, Post Graduate Centre, Kadur, Chikkamagaluru, Karnataka 577548, India
| | - K S Manjunatha
- Department of Pharmaceutical Chemistry, Kuvempu University, Post Graduate Centre, Kadur, Chikkamagaluru, Karnataka 577548, India
| | - Pooja S Rajaput
- Laboratory of Experimental Medicine, Department of Biotechnology, Kuvempu University, Shankargatta, Shimoga, Karnataka 577451, India
| | - H M Kumaraswamy
- Laboratory of Experimental Medicine, Department of Biotechnology, Kuvempu University, Shankargatta, Shimoga, Karnataka 577451, India
| | - N D Satyanarayan
- Department of Pharmaceutical Chemistry, Kuvempu University, Post Graduate Centre, Kadur, Chikkamagaluru, Karnataka 577548, India.
| |
Collapse
|
12
|
Amri F, Koulali H, Jabi R, Zazour A, Bouziane M, Ismaili Z, Kharrasse G. Pancreatic cancer: experience from an emerging country in North Africa. J Cancer Res Clin Oncol 2023; 149:14297-14302. [PMID: 37561209 DOI: 10.1007/s00432-023-05245-5] [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: 06/13/2023] [Accepted: 08/02/2023] [Indexed: 08/11/2023]
Abstract
PURPOSE Pancreatic cancer is a major global health problem, it's a highly aggressive and often has a poor prognosis. The aim of this work is to carry out a retrospective epidemiological study on pancreatic cancer in a university hospital in a North African country. METHODS This is a monocentric, descriptive and analytical retrospective cohort study carried out in the Gastroenterology department of the Mohammed VI University Hospital of Oujda, Morocco, between January 2018 and December 2022. Analysis were performed using IBM SPSS Version 21.0 RESULTS: During this period, 197 cases of pancreatic cancers were collected, the median age was 64.6 years, the majority of patients (90.9%) were over 50 year's old. and there was no significant difference in gender distribution. Among the patients, several risk factors were prevalent, including chronic tobacco smoking (22.8%), alcohol consumption (12.7%), diabetes (38.1%), obesity (7.6%), and a family history of pancreatic cancer (3%). The most common symptoms at presentation were abdominal pain and obstructive jaundice. Imaging revealed that the majority of cases were located in the head of the pancreas (80.7%). Only 19.8% of the cancers were diagnosed at a resectable stage and adenocarcinoma was the predominant histological type (96.4%). Patients with advanced cancer stages showed higher levels of CA 19-9 and hypoalbuminemia. CONCLUSION Our study aligns with previous research, indicating an increased incidence of pancreatic cancer among elderly individuals, particularly those with several risk factors such as chronic tobacco smoking, alcohol consumption, diabetes. A small proportion of cancer cases are deemed resectable at the time of diagnosis.
Collapse
Affiliation(s)
- Fakhrddine Amri
- Department of Hepato-Gastroenterology, Mohammed VI University Hospital, Oujda, Morocco.
- Digestive Diseases Research Laboratory (DSRL), Faculty of Medicine and Pharmacy, Mohammed First University, Oujda, Morocco.
| | - Hajar Koulali
- Department of Hepato-Gastroenterology, Mohammed VI University Hospital, Oujda, Morocco
- Digestive Diseases Research Laboratory (DSRL), Faculty of Medicine and Pharmacy, Mohammed First University, Oujda, Morocco
| | - Rachid Jabi
- Department of General Surgery, Mohammed VI University Hospital, Oujda, Morocco
| | - Abdelkrim Zazour
- Department of Hepato-Gastroenterology, Mohammed VI University Hospital, Oujda, Morocco
- Digestive Diseases Research Laboratory (DSRL), Faculty of Medicine and Pharmacy, Mohammed First University, Oujda, Morocco
| | - Mohammed Bouziane
- Department of General Surgery, Mohammed VI University Hospital, Oujda, Morocco
| | - Zahi Ismaili
- Department of Hepato-Gastroenterology, Mohammed VI University Hospital, Oujda, Morocco
- Digestive Diseases Research Laboratory (DSRL), Faculty of Medicine and Pharmacy, Mohammed First University, Oujda, Morocco
| | - Ghizlane Kharrasse
- Department of Hepato-Gastroenterology, Mohammed VI University Hospital, Oujda, Morocco
- Digestive Diseases Research Laboratory (DSRL), Faculty of Medicine and Pharmacy, Mohammed First University, Oujda, Morocco
| |
Collapse
|
13
|
Chahbaoui N, Khamouli S, Alaqarbeh M, Belaidi S, Sinha L, Chtita S, Bouachrine M. Identification of novel curcumin derivatives against pancreatic cancer: a comprehensive approach integrating 3D-QSAR pharmacophore modeling, virtual screening, and molecular dynamics simulations. J Biomol Struct Dyn 2023:1-19. [PMID: 37811784 DOI: 10.1080/07391102.2023.2266502] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/27/2023] [Indexed: 10/10/2023]
Abstract
Pancreatic cancer, known as the "silent killer," poses a daunting challenge in cancer therapy. The dysregulation of the PI3Kα signaling pathway in pancreatic cancer has attracted considerable interest as a promising target for therapeutic intervention. In this regard, the use of curcumin derivatives as inhibitors of PI3Kα has emerged, providing a novel and promising avenue for developing effective treatments for this devastating disease. Computational approaches were employed to explore this potential and investigate 58 curcumin derivatives with cytotoxic activity against the Panc-1 cell line. Our approach involved ligand-based pharmacophore modeling and atom-based 3D-QSAR analysis. The resulting QSAR model derived from the best-fitted pharmacophore hypothesis (AAHRR_1) demonstrated remarkable performance with high correlation coefficients (R2) of 0.990 for the training set and 0.977 for the test set. The cross-validation coefficient (Q2) of 0.971 also validated the model's predictive power. Tropsha's recommended criteria, including the Y-randomization test, were employed to ensure its reliability. Furthermore, an enrichment study was conducted to evaluate the model's performance in identifying active compounds. AAHRR_1 was used to screen a curated PubChem database of curcumin-related compounds. Two molecules (CID156189304 and CID154728220) exhibited promising pharmacokinetic properties and higher docking scores than Alpelisib, warranting further investigation. Extensive molecular dynamics simulations provided crucial insights into the conformational dynamics within the binding site, validating their stability and behavior. These findings contribute to our understanding of the potential therapeutic effectiveness of these compounds as PI3Kα inhibitors in pancreatic cancer.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Narimene Chahbaoui
- Group of Computational and Pharmaceutical Chemistry, LMCE Laboratory, University of Biskra, Biskra, Algeria
| | - Saida Khamouli
- Group of Computational and Pharmaceutical Chemistry, LMCE Laboratory, University of Biskra, Biskra, Algeria
| | - Marwa Alaqarbeh
- Basic Science Department, Prince Al Hussein Bin Abdullah II Academy for Civil Protection, Al-Balqa Applied University, Al-Salt, Jordan
| | - Salah Belaidi
- Group of Computational and Pharmaceutical Chemistry, LMCE Laboratory, University of Biskra, Biskra, Algeria
| | - Leena Sinha
- Physics Department, University of Lucknow, Lucknow, India
| | - Samir Chtita
- Laboratory of Analytical and Molecular Chemistry, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, Casablanca, Morocco
| | - Mohammed Bouachrine
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University Moulay Ismail, Meknes, Morocco
- Superior School of Technology - Khenifra (EST-Khenifra), University of Sultan Moulay Sliman, Khenifra, Morocco
| |
Collapse
|
14
|
Chen S, Phuc PT, Nguyen P, Burton W, Lin S, Lin W, Lu CY, Hsu M, Cheng C, Hsu JC. A novel prediction model of the risk of pancreatic cancer among diabetes patients using multiple clinical data and machine learning. Cancer Med 2023; 12:19987-19999. [PMID: 37737056 PMCID: PMC10587954 DOI: 10.1002/cam4.6547] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/14/2023] [Accepted: 09/06/2023] [Indexed: 09/23/2023] Open
Abstract
INTRODUCTION Pancreatic cancer is associated with poor prognosis. Considering the increased global incidence of diabetes cases and that individuals with diabetes are considered a high-risk subpopulation for pancreatic cancer, it is critical to detect the risk of pancreatic cancer within populations of person living = with diabetes. This study aimed to develop a novel prediction model for pancreatic cancer risk among patients with diabetes, using = a real-world database containing clinical features and employing numerous artificial intelligent approach algorithms. METHODS This retrospective observational study analyzed data on patients with Type 2 diabetes from a multisite Taiwanese EMR database between 2009 and 2019. Predictors were selected in accordance with the literature review and clinical perspectives. The prediction models were constructed using machine learning algorithms such as logistic regression, linear discriminant analysis, gradient boosting machine, and random forest. RESULTS The cohort consisted of 66,384 patients. The Linear Discriminant Analysis (LDA) model generated the highest AUROC of 0.9073, followed by the Voting Ensemble and Gradient Boosting machine models. LDA, the best model, exhibited an accuracy of 84.03%, a sensitivity of 0.8611, and a specificity of 0.8403. The most significant predictors identified for pancreatic cancer risk were glucose, glycated hemoglobin, hyperlipidemia comorbidity, antidiabetic drug use, and lipid-modifying drug use. CONCLUSION This study successfully developed a highly accurate 4-year risk model for pancreatic cancer in patients with diabetes using real-world clinical data and multiple machine-learning algorithms. Potentially, our predictors offer an opportunity to identify pancreatic cancer early and thus increase prevention and invention windows to impact survival in diabetic patients.
Collapse
Affiliation(s)
- Shih‐Min Chen
- School of PharmacyTaipei Medical UniversityTaipeiTaiwan
| | - Phan Thanh Phuc
- International Ph.D. Program in Biotech and Healthcare Management, College of ManagementTaipei Medical UniversityTaipeiTaiwan
| | - Phung‐Anh Nguyen
- Clinical Data Center, Office of Data ScienceTaipei Medical UniversityTaipeiTaiwan
- Clinical Big Data Research CenterTaipei Medical University Hospital, Taipei Medical UniversityTaipeiTaiwan
- Research Center of Health Care Industry Data Science, College of ManagementTaipei Medical UniversityTaipeiTaiwan
| | - Whitney Burton
- International Ph.D. Program in Biotech and Healthcare Management, College of ManagementTaipei Medical UniversityTaipeiTaiwan
| | | | - Weei‐Chin Lin
- Section of Hematology/Oncology, Department of Medicine and Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTexasUSA
| | - Christine Y. Lu
- Department of Population MedicineHarvard Medical School and Harvard Pilgrim Health Care InstituteBostonMassachusettsUSA
- Kolling Institute, Faculty of Medicine and HealthThe University of Sydney and the Northern Sydney Local Health DistrictSydneyNew South WalesAustralia
- School of Pharmacy, Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
| | - Min‐Huei Hsu
- Clinical Data Center, Office of Data ScienceTaipei Medical UniversityTaipeiTaiwan
- Graduate Institute of Data Science, College of ManagementTaipei Medical UniversityTaipeiTaiwan
| | - Chi‐Tsun Cheng
- Research Center of Health Care Industry Data Science, College of ManagementTaipei Medical UniversityTaipeiTaiwan
| | - Jason C. Hsu
- International Ph.D. Program in Biotech and Healthcare Management, College of ManagementTaipei Medical UniversityTaipeiTaiwan
- Clinical Data Center, Office of Data ScienceTaipei Medical UniversityTaipeiTaiwan
- Clinical Big Data Research CenterTaipei Medical University Hospital, Taipei Medical UniversityTaipeiTaiwan
- Research Center of Health Care Industry Data Science, College of ManagementTaipei Medical UniversityTaipeiTaiwan
| |
Collapse
|
15
|
Ma Y, Li J, Wen L, Zhang G, Yao X. Efficacy and safety of apatinib combined with radiotherapy in the treatment of advanced pancreatic cancer: a meta-analysis. World J Surg Oncol 2023; 21:165. [PMID: 37264386 DOI: 10.1186/s12957-023-03055-0] [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: 03/12/2023] [Accepted: 05/26/2023] [Indexed: 06/03/2023] Open
Abstract
OBJECTIVE At present, pancreatic cancer (PC) has a high morbidity and mortality rate and a poor prognosis. The aim of this article was to study the efficacy and safety of apatinib combined with radiotherapy in the treatment of advanced PC. METHODS The PubMed, Cochrane Library, Embase, Wanfang, CNKI, VIP, and CBM databases were searched by computer to identify studies on the application of apatinib in patients with advanced PC. The patients in the included study were divided into an observation group (apatinib combined with radiotherapy) and a control group (radiotherapy only), and meta-analysis was performed for each outcome with Revman 5.4 software. This study was successfully registered on the PROSPERO website, and the registration number is CRD: 42,022,384,056 (available at https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=384056 ). RESULTS A total of 7 randomized controlled trials (RCTs) with 428 patients were included, including 215 in the observation group and 213 in the control group. Compared with the control group, the observation group showed a greater objective response rate [OR = 3.26, 95% CI (2.18, 4.87), P < 0.0001], disease control rate [OR = 5.04, 95% CI (3.12, 8.12), P < 0.0001], complete response rate [OR = 3.87, 95% CI (1.51, 9.88), P = 0.005], and partial response rate [OR = 2.43, 95% CI (1.63, 3.61), P < 0.001], The 1-year survival rate [OR = 2.39, 95% CI (1.15, 4.96), P < 0.05], 2-year survival rate [OR = 2.41, 95% CI (1.03, 5.61), P < 0.05], progression-free survival time [MD = 1.17, 95% CI (0.37, 1.96), P < 0.05], overall survival time [MD = 1.47, 95% CI (0.13, 2.80), P < 0.05], while the stability rate [OR = 1.14, 95% CI (0.72, 1.81), P = 0.58] and various complications were not significantly different between the two groups. CONCLUSION Apatinib combined with radiotherapy was more effective than radiotherapy alone in the treatment of advanced pancreatic cancer (PC), and apatinib had acceptable safety. However, since our study was limited by the quantity and quality of the included studies, we look forward to more large-sample, multicentre, and high-quality RCTs in the future to verify the conclusions.
Collapse
Affiliation(s)
- Yongli Ma
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, No. 49 Da Gong Street, Ganzhou, 341000, China
| | - Jinghui Li
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, No. 49 Da Gong Street, Ganzhou, 341000, China
- Gannan Medical University, No. 1 Medical College Road, Ganzhou, 341000, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No.106 Zhongshan 2Nd Road, Yuexiu District, Guangzhou, 510080, China
| | - Liang Wen
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, No. 49 Da Gong Street, Ganzhou, 341000, China
- Gannan Medical University, No. 1 Medical College Road, Ganzhou, 341000, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No.106 Zhongshan 2Nd Road, Yuexiu District, Guangzhou, 510080, China
| | - Guosheng Zhang
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, No. 49 Da Gong Street, Ganzhou, 341000, China
| | - Xueqing Yao
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, No. 49 Da Gong Street, Ganzhou, 341000, China.
- Gannan Medical University, No. 1 Medical College Road, Ganzhou, 341000, China.
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No.106 Zhongshan 2Nd Road, Yuexiu District, Guangzhou, 510080, China.
| |
Collapse
|
16
|
Olajubutu O, Ogundipe OD, Adebayo A, Adesina SK. Drug Delivery Strategies for the Treatment of Pancreatic Cancer. Pharmaceutics 2023; 15:pharmaceutics15051318. [PMID: 37242560 DOI: 10.3390/pharmaceutics15051318] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/15/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Pancreatic cancer is fast becoming a global menace and it is projected to be the second leading cause of cancer-related death by 2030. Pancreatic adenocarcinomas, which develop in the pancreas' exocrine region, are the predominant type of pancreatic cancer, representing about 95% of total pancreatic tumors. The malignancy progresses asymptomatically, making early diagnosis difficult. It is characterized by excessive production of fibrotic stroma known as desmoplasia, which aids tumor growth and metastatic spread by remodeling the extracellular matrix and releasing tumor growth factors. For decades, immense efforts have been harnessed toward developing more effective drug delivery systems for pancreatic cancer treatment leveraging nanotechnology, immunotherapy, drug conjugates, and combinations of these approaches. However, despite the reported preclinical success of these approaches, no substantial progress has been made clinically and the prognosis for pancreatic cancer is worsening. This review provides insights into challenges associated with the delivery of therapeutics for pancreatic cancer treatment and discusses drug delivery strategies to minimize adverse effects associated with current chemotherapy options and to improve the efficiency of drug treatment.
Collapse
Affiliation(s)
| | - Omotola D Ogundipe
- Department of Pharmaceutical Sciences, Howard University, Washington, DC 20059, USA
| | - Amusa Adebayo
- Department of Pharmaceutical Sciences, Howard University, Washington, DC 20059, USA
| | - Simeon K Adesina
- Department of Pharmaceutical Sciences, Howard University, Washington, DC 20059, USA
| |
Collapse
|
17
|
Araujo-Abad S, Manresa-Manresa A, Rodríguez-Cañas E, Fuentes-Baile M, García-Morales P, Mallavia R, Saceda M, de Juan Romero C. New therapy for pancreatic cancer based on extracellular vesicles. Biomed Pharmacother 2023; 162:114657. [PMID: 37023623 DOI: 10.1016/j.biopha.2023.114657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/29/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC), is the most common aggressive cancer of the pancreas. The standard care of PDAC includes tumor resection and chemotherapy, but the lack of early diagnosis and the limited response to the treatment worsens the patient's condition. In order to improve the efficiency of chemotherapy, we look for more efficient systems of drug delivery. We isolated and fully characterized small Extracellular Vesicles (EVs) from the RWP-1 cell line. Our study indicates that the direct incubation method was the most efficient loading protocol and that a minimum total amount of drug triggers an effect on tumor cells. Therefore, we loaded the small EVs with two chemotherapeutic drugs (Temozolomide and EPZ015666) by direct incubation method and the amount of drug loaded was measured by high-performance liquid chromatography (HPLC). Finally, we tested their antiproliferative effect on different cancer cell lines. Moreover, the system is highly dependent on the drug structure and therefore RWP-1 small EVsTMZ were more efficient than RWP-1 small EVsEPZ015666. RWP-1 derived small EVs represent a promising drug delivery tool that can be further investigated in preclinical studies and its combination with PRMT5 inhibitor can be potentially developed in clinical trials for the treatment of PDAC.
Collapse
Affiliation(s)
- Salomé Araujo-Abad
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, 03203 Alicante, Spain; Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda, Universidad s/n, Ed. Torregaitán, Elche, 03202 Alicante, Spain; Centro de Biotecnología, Universidad Nacional de Loja, Avda. Pio Jaramillo Alvarado s/n, Loja, 110111 Loja, Ecuador
| | - Antonio Manresa-Manresa
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, 03203 Alicante, Spain
| | - Enrique Rodríguez-Cañas
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda, Universidad s/n, Ed. Torregaitán, Elche, 03202 Alicante, Spain
| | - María Fuentes-Baile
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, 03203 Alicante, Spain
| | - Pilar García-Morales
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda, Universidad s/n, Ed. Torregaitán, Elche, 03202 Alicante, Spain
| | - Ricardo Mallavia
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda, Universidad s/n, Ed. Torregaitán, Elche, 03202 Alicante, Spain
| | - Miguel Saceda
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, 03203 Alicante, Spain; Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda, Universidad s/n, Ed. Torregaitán, Elche, 03202 Alicante, Spain
| | - Camino de Juan Romero
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, 03203 Alicante, Spain; Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda, Universidad s/n, Ed. Torregaitán, Elche, 03202 Alicante, Spain.
| |
Collapse
|
18
|
Sharma NR, Lo SK, Hendifar A, Othman MO, Patel K, Mendoza-Ladd A, Verco S, Maulhardt HA, Verco J, Wendt A, Marin A, Schmidt CM, diZerega G. Response of Locally Advanced Pancreatic Cancer to Intratumoral Injection of Large Surface Area Microparticle Paclitaxel: Initial Report of Safety and Clinical Outcome. Pancreas 2023; 52:e179-e187. [PMID: 37782888 DOI: 10.1097/mpa.0000000000002236] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
OBJECTIVES Large surface area microparticle paclitaxel (LSAM-PTX) provides an intratumoral (IT) chemotherapeutic depot. Safety, tolerability, and tumor response to IT LSAM-PTX delivered by endoscopic ultrasound-fine needle injection were evaluated in subjects with unresectable locally advanced pancreatic cancer (LAPC). METHODS Ten subjects treated in a dose escalation phase and 22 additional subjects receiving 2 injections, 4 weeks apart, of 15 mg/mL LSAM-PTX were followed for 12 months. Paclitaxel pharmacokinetics were evaluated, imaging at 3 and 6 months determined tumor response, and multiplex immunofluorescence was conducted to characterize local immune response. RESULTS Most treatment-emergent adverse events were attributed to LAPC. Plasma paclitaxel levels were negligible. Eight subjects' tumors became resectable after IT LSAM-PTX, and 5 of 6 (83%) were resected with R0. Multiplex immunofluorescence of resected tumors demonstrated increased T cells, natural killer cells, and macrophages and decreased myeloid-derived suppressor cells. Six-month disease control rate was 94%, and median overall survival was 19.7 months in the 2-injection subjects. For nonresected and resected groups, overall survival times were 18.9 and 35.2 months, respectively. CONCLUSIONS Neoadjuvant IT LSAM-PTX, in combination with SOC, was well tolerated and may provide benefits to LAPC patients, evidenced by enhanced immune response, improved disease control rate, restaging leading to surgery, and extended survival.
Collapse
Affiliation(s)
- Neil R Sharma
- From the Division of Interventional Oncology and Surgical Endoscopy, Parkview Cancer Institute, Fort Wayne, IN
| | - Simon K Lo
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Andrew Hendifar
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Mohamed O Othman
- Gastroenterology and Hepatology Section, Baylor College of Medicine Medical Center, Houston, TX
| | - Kalpesh Patel
- Gastroenterology and Hepatology Section, Baylor College of Medicine Medical Center, Houston, TX
| | - Antonio Mendoza-Ladd
- Division of Gastroenterology, Texas Tech University Health Sciences Center at El Paso, El Paso, TX
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Xiang Q, Li D, Cheng C, Xu K, Zuo C. 68Ga-HBED-CC-WL-12 PET in Diagnosing and Differentiating Pancreatic Cancers in Murine Models. Pharmaceuticals (Basel) 2023; 16:80. [PMID: 36678577 PMCID: PMC9865957 DOI: 10.3390/ph16010080] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
Positron emission tomography (PET) has been proven as an important technology to detect the expression of programmed death ligand 1 (PD-L1) non-invasively and in real time. As a PD-L1 inhibitor, small peptide WL12 has shown great potential in serving as a targeting molecule to guide PD-L1 blockade therapy in clinic. In this study, WL12 was modified with HBED-CC to label 68Ga in a modified procedure, and the biologic properties were evaluated in vitro and in vivo. 68Ga-HBED-CC-WL12 showed good stability in saline and can specifically target PD-L1-positive cells U87MG and PANC02. In PANC02-bearing mice, 68Ga-HBED-CC-WL12 showed fast permeation in subcutaneous tumors within 20 min (SUVmax 0.37) and was of higher uptake in 90 min (SUVmax 0.38). When compared with 18F-FDG, 68Ga-FAPI-04, and 68Ga-RGD, 68Ga-HBED-CC-WL12 also demonstrated great image quality and advantages in evaluating immune microenvironment. This study modified the 68Ga-labeling procedure of WL12 and obtained better biologic properties and further manifested the clinical potential of 68Ga-HBED-CC-WL12 for PET imaging and guiding for immunotherapy.
Collapse
Affiliation(s)
- Qiying Xiang
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
| | - Danni Li
- Department of Nuclear Medicine, The First Affiliated Hospital (Changhai Hospital) of Naval Medical University, Shanghai 200433, China
| | - Chao Cheng
- Department of Nuclear Medicine, The First Affiliated Hospital (Changhai Hospital) of Naval Medical University, Shanghai 200433, China
| | - Kai Xu
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
| | - Changjing Zuo
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
- Department of Nuclear Medicine, The First Affiliated Hospital (Changhai Hospital) of Naval Medical University, Shanghai 200433, China
| |
Collapse
|
20
|
Qian C, Wan L, Wu Y. Analysis of the results of high-intensity focused ultrasound for patients with advanced pancreatic cancer. Int J Hyperthermia 2023; 40:2250586. [PMID: 37641497 DOI: 10.1080/02656736.2023.2250586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/18/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023] Open
Abstract
OBJECTIVE To investigate the safety, local ablation efficacy, analgesic effects, and factors influencing the survival of patients with advanced pancreatic cancer treated with high-intensity focused ultrasound (HIFU). MATERIALS AND METHODS Patients with advanced pancreatic cancer who underwent HIFU for the first time at the Suining Central Hospital between January 2018 and September 2022 were enrolled. The efficacy of tumor ablation was assessed using enhanced computed tomography (CT) and magnetic resonance imaging (MRI), pain relief was assessed using the visual analog scale (VAS), and complications and survival rates were investigated. The Kaplan-Meier method and a Cox regression model were used to analyze the independent risk factors that may have affected prognosis. RESULTS Intraoperative ultrasonography showed varying degrees of grayscale changes in all cases. One month after surgery, enhanced computed tomography or magnetic resonance imaging examinations showed complete or partial responses in 85.22% of the patients. Pain relief was achieved in 98.21% of the patients. No postoperative complications of SIR-C grade or higher were observed. The overall median survival time (MST) was 12.1 months. Cox multifactorial analysis showed that the main factors affecting overall survival (OS) were clinical stage, preoperative liver function, and combination chemotherapy. CONCLUSION HIFU is safe and effective for pancreatic cancer treatment, and has the potential to become an important supplement for the treatment of advanced pancreatic cancer. This approach needs to be further verified by multi-center and large-sample studies.
Collapse
Affiliation(s)
- Chuan Qian
- Graduate School, Zunyi Medical University, Zunyi, Guizou, China
- Department of Surgery, Suining Central Hospital, Suining, Sichuan, China
| | - LiIi Wan
- Department of Surgery, Suining Central Hospital, Suining, Sichuan, China
| | - Yakun Wu
- Graduate School, Zunyi Medical University, Zunyi, Guizou, China
- Department of Surgery, Suining Central Hospital, Suining, Sichuan, China
| |
Collapse
|
21
|
LRRC8A Is a Promising Prognostic Biomarker and Therapeutic Target for Pancreatic Adenocarcinoma. Cancers (Basel) 2022; 14:cancers14225526. [PMID: 36428619 PMCID: PMC9688930 DOI: 10.3390/cancers14225526] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/18/2022] [Accepted: 11/03/2022] [Indexed: 11/12/2022] Open
Abstract
Pancreatic adenocarcinoma (PAAD) is a highly malignant tumor of the digestive system with increasing morbidity and mortality. The lack of sensitive and reliable biomarkers is one of the main reasons for the poor prognosis. Volume-regulated anion channels (VRAC), which are ubiquitously expressed in the vertebrate cell membrane, are composed of leucine-rich repeat-containing 8A (LRRC8A) and four other homologous family members (LRRC8B-E). VRAC heterogeneous complex is implicated in each of the six "hallmarks of cancer" and represents a novel therapeutic target for cancer. In this study, LRRC8A was speculated to be a promising prognostic biomarker and therapeutic target for PAAD based on a series of bioinformatics analyses. Additional cell experiments and immunohistochemical assays demonstrated that LRRC8A can affect the prognosis of PAAD and is correlated to cell proliferation, cell migration, drug resistance, and immune infiltration. Functional analysis indicated that LRRC8A influences the progression and prognosis of patients with PAAD by the regulation of CD8+ T cells immune infiltration. Taken together, these results can help in the design of new therapeutic drugs for patients with PAAD.
Collapse
|