Advanced Cancer Diagnosis and Treatment

A special issue of Biomedicines (ISSN 2227-9059). This special issue belongs to the section "Cancer Biology and Oncology".

Deadline for manuscript submissions: closed (29 February 2024) | Viewed by 12675

Special Issue Editors


E-Mail Website
Guest Editor
Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan
Interests: cancer biodiagnostic; early detection of cancers; cancer biomarkers; neurobiology of olfaction; nematode biology
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Department of Frontier Science for Cancer and Chemotherapy, Osaka University, Suita, Japan
Interests: pancreatic cancer; gastrointestinal cancer
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Cancer is the leading cause of death worldwide. The chances of survival increase with early cancer detection, but around half of all cancers are diagnosed at an advanced stage. Significant endeavors have been made to comprehend cancer's mechanisms, create accurate and sensitive diagnostic options, and produce efficient treatments. In the last decade, advancements have been made in enhancing cancer screening programs and introducing innovative technologies to improve the early detection of cancers, with a particular focus on non-invasive methods. Novel approaches in cancer treatment are opening a new avenue for effective therapies.

Acknowledging the significant efforts made in combatting cancer, from academia to the private sector, in this Special Issue, we are seeking submissions of original research articles or reviews that present impactful advances in the development of cancer biology, diagnostics, and therapeutics on one or more of, but not limited to, the following topics:

  • Cancer biology;
  • Molecular mechanisms of cancers;
  • Cancer epigenetics;
  • Mechanism of resistance to conventional therapies;
  • Cancer biomarkers;
  • Early detection of cancers;
  • Clinical investigations;
  • Diagnostics of cancers;
  • Novel therapeutics;
  • Novel paradigms in the diagnosis and treatment of cancers.

Dr. Takaaki Hirotsu
Prof. Dr. Hideshi Ishii
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Biomedicines is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2600 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • cancer biology
  • epigenetics
  • early detection
  • diagnosis
  • biodiagnosis
  • novel therapeutics
  • new paradigms

Published Papers (10 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

19 pages, 9688 KiB  
Article
Combinatorial Therapy: Targeting CD133+ Glioma Stem-like Cells with a Polysaccharide–Prodrug Complex Functionalised Gold Nanocages
by Sreejith Raveendran, Amit Giram, Mehrnaz Elmi, Santanu Ray, Christopher Ireson, Mo Alavijeh and Irina N. Savina
Biomedicines 2024, 12(5), 934; https://doi.org/10.3390/biomedicines12050934 - 23 Apr 2024
Viewed by 557
Abstract
Cancer treatments are advancing to harness the body’s immune system against tumours, aiming for lasting effects. This progress involves combining potent chemotherapy drugs with immunogens to kill cancer cells and trigger lasting immunity. Developing new prodrugs that integrate both chemotherapy and immune-boosting elements [...] Read more.
Cancer treatments are advancing to harness the body’s immune system against tumours, aiming for lasting effects. This progress involves combining potent chemotherapy drugs with immunogens to kill cancer cells and trigger lasting immunity. Developing new prodrugs that integrate both chemotherapy and immune-boosting elements could significantly improve anticancer outcomes by activating multiple mechanisms to kill cancer cells. While bacterial polysaccharides are typically not used in therapy due to their immune-stimulating properties, we propose a safe application of an extremophilic bacterial polysaccharide, Mauran (MR), modified with the anticancer drug 5-fluorouracil (5FU) to create a novel prodrug. This obtained prodrug, chloracetyl-MR-5FU, is specifically targeted using gold nanocages to CD133+ glioma cells. Test results have shown a high encapsulation efficiency of the drug during the polysaccharide modification process; its anticancer activity was demonstrated in vitro and the release of the prodrug was demonstrated in ex vivo studies. Full article
(This article belongs to the Special Issue Advanced Cancer Diagnosis and Treatment)
Show Figures

Figure 1

14 pages, 3124 KiB  
Article
Metabolomic Approach to Identify Potential Biomarkers in KRAS-Mutant Pancreatic Cancer Cells
by Boyun Kim and Jewon Jung
Biomedicines 2024, 12(4), 865; https://doi.org/10.3390/biomedicines12040865 - 15 Apr 2024
Viewed by 635
Abstract
Pancreatic cancer is characterized by its high mortality rate and limited treatment options, often driven by oncogenic RAS mutations. In this study, we investigated the metabolomic profiles of pancreatic cancer cells based on their KRAS genetic status. Utilizing both KRAS-wildtype BxPC3 and KRAS-mutant [...] Read more.
Pancreatic cancer is characterized by its high mortality rate and limited treatment options, often driven by oncogenic RAS mutations. In this study, we investigated the metabolomic profiles of pancreatic cancer cells based on their KRAS genetic status. Utilizing both KRAS-wildtype BxPC3 and KRAS-mutant PANC1 cell lines, we identified 195 metabolites differentially altered by KRAS status through untargeted metabolomics. Principal component analysis and hierarchical condition trees revealed distinct separation between KRAS-wildtype and KRAS-mutant cells. Metabolite set enrichment analysis highlighted significant pathways such as homocysteine degradation and taurine and hypotaurine metabolism. Additionally, lipid enrichment analysis identified pathways including fatty acyl glycosides and sphingoid bases. Mapping of identified metabolites to KEGG pathways identified nine significant metabolic pathways associated with KRAS status, indicating diverse metabolic alterations in pancreatic cancer cells. Furthermore, we explored the impact of TRPML1 inhibition on the metabolomic profile of KRAS-mutant pancreatic cancer cells. TRPML1 inhibition using ML-SI1 significantly altered the metabolomic profile, leading to distinct separation between vehicle-treated and ML-SI1-treated PANC1 cells. Metabolite set enrichment analysis revealed enriched pathways such as arginine and proline metabolism, and mapping to KEGG pathways identified 17 significant metabolic pathways associated with TRPML1 inhibition. Interestingly, some metabolites identified in PANC1 compared to BxPC3 were oppositely regulated by TRPML1 inhibition, suggesting their potential as biomarkers for KRAS-mutant cancer cells. Overall, our findings shed light on the distinct metabolite changes induced by both KRAS status and TRPML1 inhibition in pancreatic cancer cells, providing insights into potential therapeutic targets and biomarkers for this deadly disease. Full article
(This article belongs to the Special Issue Advanced Cancer Diagnosis and Treatment)
Show Figures

Figure 1

17 pages, 3692 KiB  
Article
Involvement of Protease-Activated Receptor2 Pleckstrin Homology Binding Domain in Ovarian Cancer: Expression in Fallopian Tubes and Drug Design
by Jeetendra Kumar Nag, Sorina Grisaru-Granovsky, Shunit Armon, Tatyana Rudina, Priyanga Appasamy and Rachel Bar-Shavit
Biomedicines 2024, 12(1), 246; https://doi.org/10.3390/biomedicines12010246 - 22 Jan 2024
Cited by 1 | Viewed by 946
Abstract
Studying primordial events in cancer is pivotal for identifying predictive molecular indicators and for targeted intervention. While the involvement of G-protein-coupled receptors (GPCRs) in cancer is growing, GPCR-based therapies are yet rare. Here, we demonstrate the overexpression of protease-activated receptor 2 (PAR2), a [...] Read more.
Studying primordial events in cancer is pivotal for identifying predictive molecular indicators and for targeted intervention. While the involvement of G-protein-coupled receptors (GPCRs) in cancer is growing, GPCR-based therapies are yet rare. Here, we demonstrate the overexpression of protease-activated receptor 2 (PAR2), a GPCR member in the fallopian tubes (FTs) of high-risk BRCA carriers as compared to null in healthy tissues of FT. FTs, the origin of ovarian cancer, are known to express genes of serous tubal intraepithelial carcinoma (STICs), a precursor lesion of high-grade serous carcinoma (HGSC). PAR2 expression in FTs may serve as an early prediction sensor for ovarian cancer. We show now that knocking down Par2 inhibits ovarian cancer peritoneal dissemination in vivo, pointing to the central role of PAR2. Previously we identified pleckstrin homology (PH) binding domains within PAR1,2&4 as critical sites for cancer-growth. These motifs associate with PH-signal proteins via launching a discrete signaling network in cancer. Subsequently, we selected a compound from a library of backbone cyclic peptides generated toward the PAR PH binding motif, namely the lead compound, Pc(4-4). Pc(4-4) binds to the PAR PH binding domain and blocks the association of PH-signal proteins, such as Akt or Etk/Bmx with PAR2. It attenuates PAR2 oncogenic activity. The potent inhibitory function of Pc(4-4) is demonstrated via inhibition of ovarian cancer peritoneal spread in mice. While the detection of PAR2 may serve as a predictor for ovarian cancer, the novel Pc(4-4) compound may serve as a powerful medicament in STICs and ovarian cancer. This is the first demonstration of the involvement of PAR PH binding motif signaling in ovarian cancer and Pc(4-4) as a potential therapy treatment. Full article
(This article belongs to the Special Issue Advanced Cancer Diagnosis and Treatment)
Show Figures

Figure 1

13 pages, 1050 KiB  
Article
The Intraoperative Administration of Dexmedetomidine Alleviates Postoperative Inflammatory Response in Patients Undergoing Laparoscopy-Assisted Gastrectomy: A Double-Blind Randomized Controlled Trial
by Jiae Moon, Duk-Hee Chun, Hee Jung Kong, Hye Sun Lee, Soyoung Jeon, Jooeun Park, Na Young Kim and Hyoung-Il Kim
Biomedicines 2023, 11(12), 3253; https://doi.org/10.3390/biomedicines11123253 - 8 Dec 2023
Viewed by 888
Abstract
Surgical stress can compromise the immune system of patients with cancer, affecting susceptibility to perioperative infections, tumor progression, treatment responses, and postoperative recovery. Perioperatively reducing inflammatory responses could improve outcomes. We determined the impact of intraoperative dexmedetomidine administration on the inflammatory response and [...] Read more.
Surgical stress can compromise the immune system of patients with cancer, affecting susceptibility to perioperative infections, tumor progression, treatment responses, and postoperative recovery. Perioperatively reducing inflammatory responses could improve outcomes. We determined the impact of intraoperative dexmedetomidine administration on the inflammatory response and postoperative recovery in patients undergoing elective laparoscopy-assisted gastrectomy. These patients were randomly assigned to the dexmedetomidine or control group (n = 42 each). The primary endpoint was the C-reactive protein (CRP) level on postoperative day 1. The secondary endpoints included the perioperative interleukin (IL)-6 levels, postoperative numerical rating scale (NRS) scores, and rescue analgesic doses. There were no significant between-group differences in terms of CRP levels. The IL-6 levels at the end of the surgery, NRS scores in the post-anesthesia care unit, and rescue pethidine requirements within the first hour postoperatively were significantly lower in the dexmedetomidine group than in the control group. The bolus deliveries-to-attempts ratio (via patient-controlled analgesia) at 2 h differed significantly between the two groups. However, IL-6 reduction was confined to a single timepoint, and the postoperative analgesic effects lasted for the first 2 h postoperatively. Low-dose dexmedetomidine infusion (0.4 µg kg−1 h−1) during laparoscopy-assisted gastrectomy exerts minimal anti-inflammatory effects. Full article
(This article belongs to the Special Issue Advanced Cancer Diagnosis and Treatment)
Show Figures

Figure 1

15 pages, 3804 KiB  
Article
Comparative Analysis of Diagnostic Performance of Automatic Breast Ultrasound, Full-Field Digital Mammography and Contrast-Enhanced Mammography in Relation to Breast Composition
by Marta Ewa Pawlak, Wojciech Rudnicki, Anna Borkowska, Karolina Skubisz, Rafał Rydzyk and Elżbieta Łuczyńska
Biomedicines 2023, 11(12), 3226; https://doi.org/10.3390/biomedicines11123226 - 6 Dec 2023
Viewed by 861
Abstract
This single center study includes a comparative analysis of the diagnostic performance of full-field digital mammography (FFDM), contrast-enhanced mammography (CEM) and automatic breast ultrasound (ABUS) in the group of patients with breast American College of Radiology (ACR) categories C and D as well [...] Read more.
This single center study includes a comparative analysis of the diagnostic performance of full-field digital mammography (FFDM), contrast-enhanced mammography (CEM) and automatic breast ultrasound (ABUS) in the group of patients with breast American College of Radiology (ACR) categories C and D as well as A and B with FFDM. The study involved 297 patients who underwent ABUS and FFDM. Breast types C and D were determined in 40% of patients with FFDM and low- energy CEM. CEM was performed on 76 patients. Focal lesions were found in 131 patients, of which 115 were histopathologically verified. The number of lesions detected in patients with multiple lesions were 40 from 48 with ABUS, 13 with FFDM and 21 with CEM. Compliance in determining the number of foci was 82% for FFDM and 91% for both CEM and ABUS. In breast types C and D, 72% of all lesions were found with ABUS, 56% with CEM and 29% with FFDM (p = 0.008, p = 0.000); all invasive cancers were diagnosed with ABUS, 83% with CEM and 59% with FFDM (p = 0.000, p = 0.023); 100% DCIS were diagnosed with ABUS, 93% with CEM and 59% with FFDM. The size of lesions from histopathology in breast ACR categories A and B was 14–26 mm, while in breast categories C and D was 11–37 mm. In breast categories C and D, sensitivity of ABUS, FFDM and CEM was, respectively, 78.05, 85.37, 92.68; specificity: 40, 13.33, 8.33; PPV (positive predictive value): 78.05, 72.92, 77.55; NPV (negative predictive value): 40, 25, 25, accuracy: 67.86, 66.07, 73.58. In breast categories A and B, sensitivity of ABUS, FFDM and CEM was, respectively, 81.25, 93.75, 93.48; specificity: 18.18, 18.18, 16.67; PPV: 81.25, 83.33, 89.58; NPV: 18.18, 40, 25; accuracy: 69.49, 79.66, 84.62. The sensitivity of the combination of FFDM and ABUS was 100 for all types of breast categories; the accuracy was 75 in breast types C and D and 81.36 in breast types A and B. The study confirms the predominance of C and D breast anatomy types and the low diagnostic performance of FFDM within that group and indicates ABUS and CEM as potential additive methods in breast cancer diagnostics. Full article
(This article belongs to the Special Issue Advanced Cancer Diagnosis and Treatment)
Show Figures

Figure 1

20 pages, 4110 KiB  
Article
Intracellular Major Histocompatibility Complex Class II and C-X-C Motif Chemokine Ligand 10-Expressing Neutrophils Indicate the State of Anti-Tumor Activity Induced by Bacillus Calmette–Guérin
by Yuji Takeda, Tomoyuki Kato, Saima Sabrina, Sei Naito, Hiromi Ito, Naoto Emi, Yuya Kuboki, Yuki Takai, Hiroki Fukuhara, Masaki Ushijima, Takafumi Narisawa, Mayu Yagi, Hidenori Kanno, Toshihiko Sakurai, Hayato Nishida, Akemi Araki, Yoshitaka Shimotai, Mikako Nagashima, Yusuke Nouchi, Shinichi Saitoh, Hidetoshi Nara, Norihiko Tsuchiya and Hironobu Asaoadd Show full author list remove Hide full author list
Biomedicines 2023, 11(11), 3062; https://doi.org/10.3390/biomedicines11113062 - 15 Nov 2023
Cited by 2 | Viewed by 926
Abstract
(1) Background: Inflammatory responses induce the formation of both anti-tumor and pro-tumor neutrophils known as myeloid-derived suppressor cells (MDSCs). Intermittent intravesical infusion of Bacillus Calmette–Guérin (BCG) is an established cancer immunotherapy for non-muscle-invasive bladder cancer (NMIBC). However, the types of neutrophils induced via [...] Read more.
(1) Background: Inflammatory responses induce the formation of both anti-tumor and pro-tumor neutrophils known as myeloid-derived suppressor cells (MDSCs). Intermittent intravesical infusion of Bacillus Calmette–Guérin (BCG) is an established cancer immunotherapy for non-muscle-invasive bladder cancer (NMIBC). However, the types of neutrophils induced via the inflammatory response to both tumor-bearing and BCG remain unclear. (2) Methods: We therefore analyzed neutrophil dynamics in the peripheral blood and urine of patients with NMIBC who received BCG therapy. Further, we analyzed the effects of BCG in a mouse intraperitoneal tumor model. (3) Results: BCG therapy induced the formation of CXCL10 and MHC class II-positive neutrophils in the urine of patients with NMIBC but did not induce MDSC formation. CXCL10- and MHC class II-expressing neutrophils were detected in peritoneal exudate cells formed after BCG administration. Partial neutrophil depletion using an anti-Ly6G antibody suppressed the upregulation of CXCL10 and MHC class II in neutrophils and reversed the anti-tumor activity of BCG in mouse models. (4) Conclusions: These results indicated that intracellular MHC class II- and CXCL10-expressing neutrophils indicate the state of anti-tumor activity induced via BCG. The status of neutrophils in mixed inflammation of immunosuppressive and anti-tumor responses may therefore be useful for evaluating immunological systemic conditions. Full article
(This article belongs to the Special Issue Advanced Cancer Diagnosis and Treatment)
Show Figures

Figure 1

12 pages, 1779 KiB  
Article
Comparison of Efficacy and Safety of Anti-Programmed Cell Death-1 Antibody Plus Lenvatinib and Chemotherapy as First-Line Therapy for Patients with Stage IV Gallbladder Cancer: A Real-World Study in a Chinese Population
by Tiantian Wu, Changsheng Pu, Qiang Wang and Keming Zhang
Biomedicines 2023, 11(11), 2933; https://doi.org/10.3390/biomedicines11112933 - 30 Oct 2023
Viewed by 830
Abstract
Background: The present study aimed to evaluate and compare the efficacy and safety of anti-programmed cell death protein 1 (anti-PD-1) antibody plus lenvatinib (tyrosine kinase inhibitor) therapy and chemotherapy as the first-line treatment to unresectable stage IV gallbladder cancer (GBC). Methods: We retrospectively [...] Read more.
Background: The present study aimed to evaluate and compare the efficacy and safety of anti-programmed cell death protein 1 (anti-PD-1) antibody plus lenvatinib (tyrosine kinase inhibitor) therapy and chemotherapy as the first-line treatment to unresectable stage IV gallbladder cancer (GBC). Methods: We retrospectively analyzed the clinical data of patients with stage IV GBC who received chemotherapy or anti-PD-1 antibody combined with lenvatinib therapy at our hospital from March 2018 to October 2022. Patients with previous antitumor treatment were excluded. The overall survival (OS), progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR), and adverse events (AEs) were assessed. Results: A total of 64 patients were enrolled, of which 33 patients received chemotherapy (gemcitabine + cisplatin) in the chemotherapy group, and 31 patients received anti-PD-1antibody (camrelizumab) combined with lenvatinib therapy in the combined therapy group. The median OS was 12.00 months in the combined therapy group and 10.00 months in the chemotherapy group (hazard ratio (HR), 0.57; 95% CI: 0.32–1.03; p < 0.05). The median PFS was 9.00 months in the combined therapy group and 6.00 months in the chemotherapy group (HR, 0.46; 95% CI: 0.25–0.84; p < 0.01). The ORR was 54.84% and 39.39% in the combined therapy and chemotherapy groups, respectively, and the difference was not significant (p = 0.22). The DCR was 80.65% and 72.72% in the combined therapy and chemotherapy groups, respectively (p = 0.46). One patient successfully underwent radical surgery after 8 months of combined therapy and achieved a pathological complete response. Furthermore, no patients experienced AEs of hematologic toxic effects in the combined therapy group compared with the chemotherapy group, demonstrating the advantage of the combined therapy. Conclusions: Anti-PD-1 antibody combined with lenvatinib may be a potentially effective and tolerable first-line treatment for unresectable stage IV GBC. Full article
(This article belongs to the Special Issue Advanced Cancer Diagnosis and Treatment)
Show Figures

Figure 1

15 pages, 2442 KiB  
Article
Regulation of 4-HNE via SMARCA4 Is Associated with Worse Clinical Outcomes in Hepatocellular Carcinoma
by Shiori Watabe, Yukari Aruga, Ryoko Kato, Genji Kawade, Yuki Kubo, Anna Tatsuzawa, Iichiroh Onishi, Yuko Kinowaki, Sachiko Ishibashi, Masumi Ikeda, Yuki Fukawa, Keiichi Akahoshi, Minoru Tanabe, Morito Kurata, Kenichi Ohashi, Masanobu Kitagawa and Kouhei Yamamoto
Biomedicines 2023, 11(8), 2278; https://doi.org/10.3390/biomedicines11082278 - 16 Aug 2023
Cited by 1 | Viewed by 1941
Abstract
Accumulation of 4-hydroxynonenal (4-HNE), a marker of lipid peroxidation, has various favorable and unfavorable effects on cancer cells; however, the clinicopathological significance of its accumulation in hepatocellular carcinoma (HCC) and its metabolic pathway remain unknown. This study analyzed 4-HNE accumulation and its clinicopathological [...] Read more.
Accumulation of 4-hydroxynonenal (4-HNE), a marker of lipid peroxidation, has various favorable and unfavorable effects on cancer cells; however, the clinicopathological significance of its accumulation in hepatocellular carcinoma (HCC) and its metabolic pathway remain unknown. This study analyzed 4-HNE accumulation and its clinicopathological significance in HCC. Of the 221 cases, 160 showed relatively low accumulation of 4-HNE in HCC tissues, which was an independent prognostic predictor. No correlation was found between 4-HNE accumulation and the expression of the antioxidant enzymes glutathione peroxidase 4, ferroptosis suppressor protein 1, and guanosine triphosphate cyclohydrolase 1. Therefore, we hypothesized that 4-HNE metabolism is up-regulated in HCC. A database search was focused on the transcriptional regulation of aldo-keto reductases, alcohol dehydrogenases, and glutathione-S-transferases, which are the metabolic enzymes of 4-HNE, and seven candidate transcription factor genes were selected. Among the candidate genes, the knockdown of SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a, member 4 (SMARCA4) increased 4-HNE accumulation. Immunohistochemical analysis revealed an inverse correlation between 4-HNE accumulation and SMARCA4 expression. These results suggest that SMARCA4 regulates 4-HNE metabolism in HCC. Therefore, targeting SMARCA4 provides a basis for a new therapeutic strategy for HCC via 4-HNE accumulation and increased cytotoxicity. Full article
(This article belongs to the Special Issue Advanced Cancer Diagnosis and Treatment)
Show Figures

Figure 1

Review

Jump to: Research

30 pages, 3155 KiB  
Review
Pancreatic Cancer and Detection Methods
by Umbhorn Ungkulpasvich, Hideyuki Hatakeyama, Takaaki Hirotsu and Eric di Luccio
Biomedicines 2023, 11(9), 2557; https://doi.org/10.3390/biomedicines11092557 - 18 Sep 2023
Viewed by 3123
Abstract
The pancreas is a vital organ with exocrine and endocrine functions. Pancreatitis is an inflammation of the pancreas caused by alcohol consumption and gallstones. This condition can heighten the risk of pancreatic cancer (PC), a challenging disease with a high mortality rate. Genetic [...] Read more.
The pancreas is a vital organ with exocrine and endocrine functions. Pancreatitis is an inflammation of the pancreas caused by alcohol consumption and gallstones. This condition can heighten the risk of pancreatic cancer (PC), a challenging disease with a high mortality rate. Genetic and epigenetic factors contribute significantly to PC development, along with other risk factors. Early detection is crucial for improving PC outcomes. Diagnostic methods, including imagining modalities and tissue biopsy, aid in the detection and analysis of PC. In contrast, liquid biopsy (LB) shows promise in early tumor detection by assessing biomarkers in bodily fluids. Understanding the function of the pancreas, associated diseases, risk factors, and available diagnostic methods is essential for effective management and early PC detection. The current clinical examination of PC is challenging due to its asymptomatic early stages and limitations of highly precise diagnostics. Screening is recommended for high-risk populations and individuals with potential benign tumors. Among various PC screening methods, the N-NOSE plus pancreas test stands out with its high AUC of 0.865. Compared to other commercial products, the N-NOSE plus pancreas test offers a cost-effective solution for early detection. However, additional diagnostic tests are required for confirmation. Further research, validation, and the development of non-invasive screening methods and standardized scoring systems are crucial to enhance PC detection and improve patient outcomes. This review outlines the context of pancreatic cancer and the challenges for early detection. Full article
(This article belongs to the Special Issue Advanced Cancer Diagnosis and Treatment)
Show Figures

Figure 1

13 pages, 1585 KiB  
Review
Drug Discovery and Development of miRNA-Based Nucleotide Drugs for Gastrointestinal Cancer
by Hiromichi Sato, Tomoaki Hara, Sikun Meng, Yoshiko Tsuji, Yasuko Arao, Kazuki Sasaki, Norikatsu Miyoshi, Shogo Kobayashi, Yuichiro Doki, Hidetoshi Eguchi and Hideshi Ishii
Biomedicines 2023, 11(8), 2235; https://doi.org/10.3390/biomedicines11082235 - 9 Aug 2023
Cited by 1 | Viewed by 1196
Abstract
Short non-coding RNAs, miRNAs, play roles in the control of cell growth and differentiation in cancer. Reportedly, the introduction of miRNAs could reduce the biologically malignant behavior of cancer cells, suggesting a possible use as therapeutic reagents. Given that the forced expression of [...] Read more.
Short non-coding RNAs, miRNAs, play roles in the control of cell growth and differentiation in cancer. Reportedly, the introduction of miRNAs could reduce the biologically malignant behavior of cancer cells, suggesting a possible use as therapeutic reagents. Given that the forced expression of several miRNAs, including miR-302, results in the cellular reprograming of human and mouse cells, which is similar to the effects of the transcription factors Oct4, Sox2, Klf4, and c-Myc, this suggests that the selective introduction of several miRNAs will be able to achieve anti-cancer effects at the epigenetic and metabolic levels. In this review article, we bring together the recent advances made in studies of microRNA-based therapeutic approaches to therapy-resistant cancers, especially in gastrointestinal organs. Full article
(This article belongs to the Special Issue Advanced Cancer Diagnosis and Treatment)
Show Figures

Figure 1

Back to TopTop