1
|
German-Cortés J, Herrero R, Torroglosa N, Pumarola A, Fischer-Albiol N, Campos-Moreno S, Sabaté S, Alcina À, Mancilla S, García B, Llaguno-Munive M, Díaz-Riascos ZV, Martins C, Schwartz S, Ferrer-Costa R, Abasolo I, Sánchez-Gómez P, Sarmento B, Rafael D, Andrade F. Preclinical evaluation of several polymeric micelles identifies Soluplus®-docetaxel as the most effective candidate in multiple glioblastoma models. J Control Release 2025; 381:113616. [PMID: 40073942 DOI: 10.1016/j.jconrel.2025.113616] [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: 11/10/2024] [Revised: 01/23/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
Glioblastoma multiforme (GBM) is one of the most lethal cancers, with limited treatment options due to the blood-brain barrier (BBB), systemic toxicity, and treatment resistance. Nanomedicine offers potential solutions to these challenges. This study explores Pluronic® F127 and Soluplus®-based micelles as carriers for Lomustine, Gefitinib, and Docetaxel to determine the optimal system for GBM therapy. Micelles were physicochemically characterized and biologically validated using U87-MG and U251-MG GBM cell lines in 2D and 3D models, assessing internalization, safety, and therapeutic efficacy. Soluplus® micelles (SM) showed favorable properties for intravenous administration, including low polydispersity, efficient drug release in the tumoral microenvironment, minimal cell toxicity, and a BBB-crossing rate of 15 %. Among the drugs tested, Docetaxel showed the lowest IC50 values in both 2D cell models and demonstrated superior efficacy in 3D cultures when delivered by SM. Molecular analysis confirmed that SM-D impacts key GBM-related pathways, affecting markers like E-cadherin, EPCAM, L1CAM, or EGFR. In vivo, SM-D significantly reduced tumor mass and cancer cell density, showing a favorable safety profile compared to free Docetaxel, as evidenced by reduced weight loss and histological assessments. Overall, SM-D stands out as the most promising approach for GBM treatment, supporting the potential of nanomedicine in overcoming the barriers to effective glioblastoma therapy.
Collapse
Affiliation(s)
- Júlia German-Cortés
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain; Doctoral program in Biotechnology, Faculty of Pharmacy and Food Sciences, School of Pharmacy, Universitat de Barcelona (UB), Av. de Joan XXIII, 27-31, 08028 Barcelona, Spain
| | - Raquel Herrero
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Natalia Torroglosa
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Alexandra Pumarola
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Narine Fischer-Albiol
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Sofia Campos-Moreno
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Sofia Sabaté
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Àngels Alcina
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain; Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
| | - Sandra Mancilla
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain; Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, Barcelona, Spain
| | - Belén García
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain; Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, Barcelona, Spain
| | - Monserrat Llaguno-Munive
- Laboratorio de Física Médica, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Av. San Fernando 22, 14080 Ciudad de México, CDMX, Mexico
| | - Zamira V Díaz-Riascos
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain; Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, Barcelona, Spain
| | - Cláudia Martins
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal
| | - Simó Schwartz
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain; Clinical Biochemistry Service, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Roser Ferrer-Costa
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain; Clinical Biochemistry Service, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Ibane Abasolo
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain; Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, Barcelona, Spain; Clinical Biochemistry Service, Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Pilar Sánchez-Gómez
- Unidad de Neurobiología Molecular, Área de Biología Celular y del Desarrollo, Instituto de Salud Carlos III, Ctra. Pozuelo-Majadahonda, km 2, 28220 Madrid, Spain
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; IUCS-CESPU, Rua Central de Gandra 1317, Gandra 4585-116, Portugal
| | - Diana Rafael
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain; Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingenería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto De Salud Carlos III, Barcelona, Spain.
| | - Fernanda Andrade
- Clinical Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d'Hebron Institut of Research (VHIR), Vall d'Hebron University Hospital, Vall d'Hebron Barcelona Hospital Campus, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain; Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; Department of Pharmacy and Pharmaceutical Technology and Physicochemistry, Faculty of Pharmacy and Food Sciences, School of Pharmacy, Universitat de Barcelona (UB), Av. de Joan XXIII, 27-31, 08028 Barcelona, Spain.
| |
Collapse
|
2
|
Fang M, Wu Z, Xia Z, Xiao J. Diagnostic, prognostic, and immunological roles of NCAPG in pan-cancer: A bioinformatics analysis. Medicine (Baltimore) 2025; 104:e41761. [PMID: 40068055 PMCID: PMC11903004 DOI: 10.1097/md.0000000000041761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 11/13/2024] [Accepted: 02/16/2025] [Indexed: 03/14/2025] Open
Abstract
Growing studies have shown that non-SMC condensin I complex subunit G (NCAPG) was highly expressed in a variety of tumors and was involved in the progression of multitumors, but the role of NCAPG in tumorigenesis is not fully understood. Our study purposed to systematically investigate the role of NCAPG across cancer types. Interacting molecules with NCAPG were analyzed using searching bioinformatics websites including Search Tool for the Retrieval of Interacting Genes/Proteins, GeneMANIA, and Global Positioning System-Prot. NCAPG-related diseases were acquired using the Open Targets Platform. The interaction of NCAPG and 14 cancer functional states was achieved using the CancerSEA website. The databases including the University of California Santa Cruz Xena, Genotype-Tissue Expression, The Cancer Genome Atlas Program, Human Protein Atlas, and XIANTAO Academic were used to interpret the expression of NCAPG. Correlations between NCAPG expression and immune infiltration and immune-related molecules were analyzed by using Tumor Immune Estimation Resource Version 2 and Tumor and Immune System Interaction Database databases. NCAPG expression was significantly upregulated in most cancer types. NCAPG was identified as a marker of diagnostic value and prognostic significance in most cancer types. NCAPG expression was related to immune cell infiltration and immune-related molecules across various cancers, especially kidney renal clear cell carcinoma and thyroid carcinoma. Furthermore, NCAPG expression could affect the enrichment and decrease immune cell infiltration to influence prognosis in kidney renal clear cell carcinoma but was devoid of evidence in thyroid carcinoma. NCAPG was a prospective marker for the diagnosis and prognosis of pan-cancer. Our results suggested that NCAPG was a potential cancer biomarker for the diagnosis and prognosis of pan-cancer. NCAPG might affect the immune microenvironment, which could be applied in the development of new-targeted drugs for immunotherapy.
Collapse
Affiliation(s)
- Min Fang
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China
- “The 14th Five-Year Plan” Application Characteristic Discipline of Hunan Province (Pharmaceutical Science) Changsha Medical University, Changsha, China
| | - Zhu Wu
- Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University, Changsha, China
| | - Zhi Xia
- Department of Oncology, Hunan Provincial People’s Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Jian Xiao
- Department of Geriatric Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
3
|
Oh C, Kim MS, Shin U, Kang JW, Kim YH, Ko HS, Ra JS, Ahn S, Choi EY, Yu S, Nam U, Choi T, Myung K, Lee Y. SMC2 and Condensin II Subunits Are Essential for the Development of Hematopoietic Stem and Progenitor Cells in Zebrafish. J Cell Physiol 2025; 240:e70023. [PMID: 40134128 PMCID: PMC11937623 DOI: 10.1002/jcp.70023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 02/27/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025]
Abstract
Hematopoietic stem and progenitor cells (HSPCs) play a pivotal role in blood cell production, maintaining the health and homeostasis of individuals. Dysregulation of HSPC function can lead to blood-related diseases, including cancer. Despite its importance, our understanding of the genes and pathways underlying HSPC development and the associated pathological mechanisms remains limited. To elucidate these unknown mechanisms, we analyzed databases of patients with blood disorders and performed functional gene studies using zebrafish. We employed bioinformatics tools to explore three public databases focusing on patients with myelodysplastic syndrome (MDS) and related model studies. This analysis identified significant alterations in several genes, especially SMC2 and other condensin-related genes, in patients with MDS. To further investigate the role of Smc2 in hematopoiesis, we generated smc2 loss-of-function zebrafish mutants using CRISPR mutagenesis. Further analyses of the mutants revealed that smc2 depletion induced G2/M cell cycle arrest in HSPCs, leading to their maintenance and expansion failure. Notably, although the condensin II subunits (ncaph2, ncapg2, and ncapd3) were essential for HSPC maintenance, the condensin I subunits did not affect HSPC development. These findings emphasize the crucial role of condensin II in ensuring healthy hematopoiesis via promoting HSPC proliferation.
Collapse
Affiliation(s)
- Chang‐Kyu Oh
- Center for Genomic Integrity, Institute for Basic ScienceUlsanRepublic of Korea
- Department of Biochemistry, School of MedicinePusan National UniversityYangsanRepublic of Korea
- Institute for Future EarthPusan National UniversityPusanRepublic of Korea
| | - Man S. Kim
- Clinical Research Institute, Kyung Hee University Hospital at GangdongKyung Hee UniversitySeoulRepublic of Korea
| | - Unbeom Shin
- Center for Genomic Integrity, Institute for Basic ScienceUlsanRepublic of Korea
| | - Ji Wan Kang
- Department of Anatomy, School of MedicinePusan National UniversityYangsanRepublic of Korea
| | - Yun Hak Kim
- Department of Anatomy, School of MedicinePusan National UniversityYangsanRepublic of Korea
- Department of Biomedical Informatics, School of MedicinePusan National UniversityYangsanRepublic of Korea
| | - Hwa Soo Ko
- Center for Genomic Integrity, Institute for Basic ScienceUlsanRepublic of Korea
| | - Jae Sun Ra
- Center for Genomic Integrity, Institute for Basic ScienceUlsanRepublic of Korea
| | - Soyul Ahn
- Department of Biochemistry, School of MedicinePusan National UniversityYangsanRepublic of Korea
- Institute for Future EarthPusan National UniversityPusanRepublic of Korea
| | - Eun Young Choi
- Department of Biochemistry, School of MedicinePusan National UniversityYangsanRepublic of Korea
| | - Sanghyeon Yu
- Department of Biomedical Science and TechnologyKyung Hee UniversitySeoulRepublic of Korea
| | - Uijeong Nam
- Department of Biomedical Science and TechnologyKyung Hee UniversitySeoulRepublic of Korea
| | - Taesoo Choi
- Department of Urology, School of MedicineKyung Hee UniversitySeoulRepublic of Korea
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic ScienceUlsanRepublic of Korea
- Department of Biomedical EngineeringUlsan National Institute for Science and TechnologyUlsanRepublic of Korea
| | - Yoonsung Lee
- Clinical Research Institute, Kyung Hee University Hospital at GangdongKyung Hee UniversitySeoulRepublic of Korea
| |
Collapse
|
4
|
Gu R, Mei K, Chen Z, Huang Y, Wang F. Development and validation of a prognosis prediction model for overall survival in correlation between butyrate metabolism and gastric cancer prognosis: Mendelian randomization and transcriptomics analysis. Transl Cancer Res 2025; 14:743-760. [PMID: 40104718 PMCID: PMC11912061 DOI: 10.21037/tcr-24-677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 12/17/2024] [Indexed: 03/20/2025]
Abstract
Background Gastric cancer (GC) remains a leading cause of cancer-related mortality due to its late diagnosis and poor prognosis. Butyrate metabolism (BM) has demonstrated significant roles in tumor biology, but its prognostic implications in GC remain unexplored. We aimed to investigate the effect of butyrate metabolic biomarkers on the prognosis of GC. Methods We acquired datasets from The Cancer Genome Atlas and Gene Expression Omnibus. Differential BM-related genes (BMGs) were identified using weighted gene co-expression network analysis (WGCNA). Patients were stratified into subtypes, and a prognostic model was constructed using least absolute shrinkage and selection operator (LASSO) regression. Mendelian randomization (MR) analysis was conducted using genetic variants as instrumental variables to establish causal links between BM and GC prognosis. Results Our model demonstrated robust prognostic accuracy with an area under the receiver operating characteristic (ROC) curve of 0.716. Transcriptomic analysis identified two key BMGs, SMC2 and HSPB1, with significant implications for GC survival. However, MR analysis provided no evidence of a causal association between BM and GC. Conclusions We identified two butyrate metabolic prognostic genes, namely, structural maintenance of chromosome 2 and heat shock protein beta-1, as the prognostic markers for GC. Furthermore, MR indicated no causal association between the butyrate metabolic pathway and GC.
Collapse
Affiliation(s)
- Renjun Gu
- Department of Gastroenterology and Hepatology, Jinling Hospital, Medical School of Nanjing University, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- School of Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Kun Mei
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Zilu Chen
- Department of Ultrasound, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Huang
- Department of Ultrasound, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Fangyu Wang
- Department of Gastroenterology and Hepatology, Jinling Hospital, Medical School of Nanjing University, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
5
|
McMahon R, Lucas N, Hill C, Pascovici D, Herbert B, Karsten E. Investigating the Use of Novel Blood Processing Methods to Boost the Identification of Biomarkers for Non-Small Cell Lung Cancer: A Proof of Concept. J Proteome Res 2025; 24:344-355. [PMID: 39642266 DOI: 10.1021/acs.jproteome.4c00829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2024]
Abstract
Diagnosis of non-small cell lung cancer (NSCLC) currently relies on imaging; however, these methods are not effective for detecting early stage disease. Investigating blood-based protein biomarkers aims to simplify the diagnostic process and identify disease-associated changes before they can be seen by using imaging techniques. In this study, plasma and frozen whole blood cell pellets from NSCLC patients and healthy controls were processed using both classical and novel techniques to produce a unique set of four sample types from a single blood draw. These samples were analyzed using 12 immunoassays and liquid chromatography-mass spectrometry to collectively screen 3974 proteins. Analysis of all fractions produced a set of 522 differentially expressed proteins, with conventional blood analysis (proteomic analysis of plasma) accounting for only 7 of the total. Boosted regression tree analysis of the differentially expressed proteins produced a panel of 13 proteins that were able to discriminate between controls and NSCLC patients, with an area under the ROC curve (AUC) of 0.864 for the set. Our rapid and reproducible (<10% CV for technical replicates) blood preparation and analysis methods enabled the production of high-quality data from only 30 μL of complex samples that typically require significant fractionation prior to proteomic analysis. With our methods, almost 4000 proteins were identified from a single fraction over a 62.5 min gradient by LC-MS/MS.
Collapse
Affiliation(s)
- Rosalee McMahon
- Sangui Bio Pty Ltd, Sydney 2065, Australia
- The Kolling Institute, Sydney 2065, Australia
| | - Natasha Lucas
- Sangui Bio Pty Ltd, Sydney 2065, Australia
- The Kolling Institute, Sydney 2065, Australia
- University of Sydney, Sydney 2050, Australia
| | - Cameron Hill
- Sangui Bio Pty Ltd, Sydney 2065, Australia
- The Kolling Institute, Sydney 2065, Australia
| | - Dana Pascovici
- Insight Stats, Sydney 2133, Australia
- Current: CSIRO Health &Biosecurity, Westmead 2145, Australia
| | - Ben Herbert
- Sangui Bio Pty Ltd, Sydney 2065, Australia
- The Kolling Institute, Sydney 2065, Australia
| | - Elisabeth Karsten
- Sangui Bio Pty Ltd, Sydney 2065, Australia
- The Kolling Institute, Sydney 2065, Australia
- University of Sydney, Sydney 2050, Australia
| |
Collapse
|
6
|
Lv H, Du Y, Xin P, Zhong Y, Liu Z, Jing Y. Identification of prevention marker associated with DNA replication in ovarian cancer: Expression of MCM2 protein and bioinformatics analysis. Int J Biol Macromol 2024; 279:135079. [PMID: 39187104 DOI: 10.1016/j.ijbiomac.2024.135079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/03/2024] [Accepted: 08/24/2024] [Indexed: 08/28/2024]
Abstract
Ovarian cancer is one of the types of gynecological cancers that is considered to be particularly dangerous. Ovarian cancer treatment has come a long way in recent years, but the disease is still quite likely to spread to other parts of the body. In this line of research, our goal is to pinpoint the shifts in gene expression profiles that are responsible for the avoidance of ovarian cancer. The dataset GSE54388 which was deposited in the Gene Expression Omnibus (GEO) database was processed in order to find differentially expressed genes (DEGs) that were present between human ovarian surface epithelium samples and tumor epithelial component samples. The weighted gene correlation network analysis, also known as WGCNA, was performed on the modules that were associated with the ovarian cancer group. The Gene Ontology (GO), the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, and the Gene Set Enrichment Analysis (GSEA) were used to compile a summary of the DEGs that were found in the Venn analysis of the Royalbule module. This analysis found 186 genes that overlapped in the royal blue module. Using the cytohubba plug-in that is included in the Cytoscape software, the Protein-protein Interaction (PPI) network was created and then searched to identify hub genes. Based on these findings, it seems that 10 genes have a role as hub genes in the prevention of ovarian cancer.
Collapse
Affiliation(s)
- Huina Lv
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Yang Du
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Peng Xin
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Yifan Zhong
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Zhe Liu
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Yuchen Jing
- Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| |
Collapse
|
7
|
Cepero A, Jiménez-Carretero M, Jabalera Y, Gago L, Luque C, Cabeza L, Melguizo C, Jimenez-Lopez C, Prados J. LGR5 as a Therapeutic Target of Antibody-Functionalized Biomimetic Magnetoliposomes for Colon Cancer Therapy. Int J Nanomedicine 2024; 19:1843-1865. [PMID: 38414530 PMCID: PMC10898605 DOI: 10.2147/ijn.s440881] [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: 10/18/2023] [Accepted: 01/03/2024] [Indexed: 02/29/2024] Open
Abstract
Purpose The lack of specificity of conventional chemotherapy is one of the main difficulties to be solved in cancer therapy. Biomimetic magnetoliposomes are successful chemotherapy controlled-release systems, hyperthermia, and active targeting agents by functionalization of their surface with monoclonal antibodies. The membrane receptor Leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5) stands out as colorectal cancer (CRC) biomarker and appears to be related to treatment resistance and the development of metastasis. The aim of this study was to assess the effectiveness and safety of LGR5-targeted biomimetic magnetoliposomes loaded with oxaliplatin (OXA) or 5-fluorouracil (5-FU) in the selective treatment of CRC and their possible application in hyperthermia. Methods Synthesis, characterization and determination of heating capacity of magnetoliposomes transporting OXA or 5-FU (with and without LGR5 functionalization) were conducted. In vitro antitumoral activity was assayed in multiple colorectal cell lines at different times of exposition. In addition to this, cell internalization was studied by Prussian Blue staining, flow cytometry and fluorescence microscopy. In vivo acute toxicity of magnetoliposomes was performed to evaluate iron-related toxicity. Results OXA and 5-FU loaded magnetoliposomes functionalized with LGR5 antibody showed higher cellular uptake than non-targeted nanoformulation with a reduction of the percentage of proliferation in colon cancer cell lines up to 3.2-fold of the IC50 value compared to that of free drug. The differences between non-targeted and targeted nanoformulations were more evident after short exposure times (4 and 8 hours). Interestingly, assays in the MC38 transduced cells with reduced LGR5 expression (MC38-L(-)), showed lower cell internalization of LGR5-targeted magnetoliposomes compared to non-transduced MC38 cell line. In addition, magnetoliposomes showed an in vitro favorable heating response under magnetic excitation and great iron-related biocompatibility data in vivo. Conclusion Drug-loaded magnetoliposomes functionalized with anti-LGR5 antibodies could be a promising CRC treatment strategy for LGR5+ targeted chemotherapy, magnetic hyperthermia, and both in combination.
Collapse
Affiliation(s)
- Ana Cepero
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain
- Biosanitary Research Institute ibs.GRANADA, Granada, 18012, Spain
| | | | - Ylenia Jabalera
- Department of Microbiology, Sciences School, University of Granada, Granada, 18002, Spain
| | - Lidia Gago
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain
- Biosanitary Research Institute ibs.GRANADA, Granada, 18012, Spain
| | - Cristina Luque
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain
- Biosanitary Research Institute ibs.GRANADA, Granada, 18012, Spain
| | - Laura Cabeza
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain
- Biosanitary Research Institute ibs.GRANADA, Granada, 18012, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain
- Biosanitary Research Institute ibs.GRANADA, Granada, 18012, Spain
| | | | - José Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain
- Biosanitary Research Institute ibs.GRANADA, Granada, 18012, Spain
| |
Collapse
|
8
|
Rafael D, Montero S, Carcavilla P, Andrade F, German-Cortés J, Diaz-Riascos ZV, Seras-Franzoso J, Llaguno M, Fernández B, Pereira A, Duran-Lara EF, Schwartz S, Abasolo I. Intracellular Delivery of Anti-Kirsten Rat Sarcoma Antibodies Mediated by Polymeric Micelles Exerts Strong In Vitro and In Vivo Anti-Tumorigenic Activity in Kirsten Rat Sarcoma-Mutated Cancers. ACS APPLIED MATERIALS & INTERFACES 2023; 15:10398-10413. [PMID: 36795046 DOI: 10.1021/acsami.2c19897] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The Kirsten rat sarcoma viral oncogene (KRAS) is one of the most well-known proto-oncogenes, frequently mutated in pancreatic and colorectal cancers, among others. We hypothesized that the intracellular delivery of anti-KRAS antibodies (KRAS-Ab) with biodegradable polymeric micelles (PM) would block the overactivation of the KRAS-associated cascades and revert the effect of its mutation. To this end, PM-containing KRAS-Ab (PM-KRAS) were obtained using Pluronic F127. The feasibility of using PM for antibody encapsulation as well as the conformational change of the polymer and its intermolecular interactions with the antibodies was studied, for the first time, using in silico modeling. In vitro, encapsulation of KRAS-Ab allowed their intracellular delivery in different pancreatic and colorectal cancer cell lines. Interestingly, PM-KRAS promoted a high proliferation impairment in regular cultures of KRAS-mutated HCT116 and MIA PaCa-2 cells, whereas the effect was neglectable in non-mutated or KRAS-independent HCT-8 and PANC-1 cancer cells, respectively. Additionally, PM-KRAS induced a remarkable inhibition of the colony formation ability in low-attachment conditions in KRAS-mutated cells. In vivo, when compared with the vehicle, the intravenous administration of PM-KRAS significantly reduced tumor volume growth in HCT116 subcutaneous tumor-bearing mice. Analysis of the KRAS-mediated cascade in cell cultures and tumor samples showed that the effect of PM-KRAS was mediated by a significant reduction of the ERK phosphorylation and a decrease in expression in the stemness-related genes. Altogether, these results unprecedently demonstrate that the delivery of KRAS-Ab mediated by PM can safely and effectively reduce the tumorigenicity and the stemness properties of KRAS-dependent cells, thus bringing up new possibilities to reach undruggable intracellular targets.
Collapse
Affiliation(s)
- Diana Rafael
- Drug Delivery & Targeting, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona 08035, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
- Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona 08035, Spain
| | - Sara Montero
- Drug Delivery & Targeting, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona 08035, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Pilar Carcavilla
- Drug Delivery & Targeting, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona 08035, Spain
| | - Fernanda Andrade
- Drug Delivery & Targeting, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona 08035, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
- Departament de Farmàcia i Tecnologia Farmacèutica i Fisicoquímica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), Barcelona 08028, Spain
| | - Júlia German-Cortés
- Drug Delivery & Targeting, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona 08035, Spain
| | - Zamira V Diaz-Riascos
- Drug Delivery & Targeting, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona 08035, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
- Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona 08035, Spain
| | - Joaquin Seras-Franzoso
- Drug Delivery & Targeting, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona 08035, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Monserrat Llaguno
- Drug Delivery & Targeting, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona 08035, Spain
| | - Begoña Fernández
- Drug Delivery & Targeting, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona 08035, Spain
| | - Alfredo Pereira
- Departamento de Química Orgánica y Fisicoquímica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Metropolitan Region 8380492, Chile
| | - Esteban F Duran-Lara
- Bio and NanoMaterials Lab, Drug Delivery and Controlled Release, Departamento de Microbiología, Facultad de Ciencias de la Salud, Universidad de Talca, P.O. Box 747, Talca, Maule 1141, Chile
- Center for Nanomedicine, Diagnostic & Drug Development (ND3), Universidad de Talca, P.O. Box 747, Talca, Maule 1141, Chile
| | - Simó Schwartz
- Drug Delivery & Targeting, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona 08035, Spain
- Servei de Bioquímica, Hospital Universitari Vall d'Hebron, Barcelona 08035, Spain
| | - Ibane Abasolo
- Drug Delivery & Targeting, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona 08035, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
- Functional Validation & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona 08035, Spain
- Servei de Bioquímica, Hospital Universitari Vall d'Hebron, Barcelona 08035, Spain
| |
Collapse
|
9
|
Mishra S, Streeter PR. Micelle-Based Nanocarriers for Targeted Delivery of Cargo to Pancreas. Methods Mol Biol 2022; 2592:175-184. [PMID: 36507993 DOI: 10.1007/978-1-0716-2807-2_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Innovations in the field of amphiphilic block copolymers have led to the development of a series of attractive polymer-based drug and gene delivery micellar formulations. The amphiphilic block copolymers' low critical micelle concentration (CMC) results in highly stable nanoscale micelles possessing favorable in vivo safety profiles and biocompatibility, making them an excellent carrier choice for the systemic administration of various poorly soluble drugs. These micelles can also be used as an actively targeted drug delivery system. The targeting is achieved by conjugating specific targeting ligand molecules to the micelle surface. The conjugation takes place at the hydrophilic termini of the copolymers, which forms the shell or surface of the nanomicelles. In our lab, we have developed a targeted Pluronic® F127-based nanoformulation to achieve targeting of specific cell types in the pancreas. To achieve active targeting based on the desired end application, we have conjugated several monoclonal antibodies (~150 kDa IgG) reactive to specific cell types in the pancreas by coupling lysine amino groups of the antibody to the p-nitrophenyl carbonate groups generated on the hydrophilic PEO segments of the Pluronic® F127. The resultant targeted nanomicelles demonstrated high binding specificity and targeting efficiency. These nanomicelles can be used to encapsulate and deliver hydrophobic imaging agents and/or water-insoluble therapeutic small molecules to specific pancreatic cell types, enabling the development of diverse tools for use in the diagnosis and/or treatment of pancreas pathologies.
Collapse
Affiliation(s)
- Swati Mishra
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA.
- Department of Pediatrics, Papé Family Pediatric Research Institute, Oregon Stem Cell Center, Oregon Health and Science University, OR, USA.
| | - Philip R Streeter
- Department of Pediatrics, Papé Family Pediatric Research Institute, Oregon Stem Cell Center, Oregon Health and Science University, OR, USA
| |
Collapse
|
10
|
Cepero A, Luque C, Cabeza L, Perazzoli G, Quiñonero F, Mesas C, Melguizo C, Prados J. Antibody-Functionalized Nanoformulations for Targeted Therapy of Colorectal Cancer: A Systematic Review. Int J Nanomedicine 2022; 17:5065-5080. [PMID: 36345508 PMCID: PMC9635983 DOI: 10.2147/ijn.s368814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 08/21/2022] [Indexed: 11/06/2022] Open
Abstract
The failure of chemotherapeutic treatment in colorectal cancer (CRC), the second most mortal cancer worldwide, is associated with several drug limitations, such as non-selective distribution, short half-life, and development of multiple resistances. One of the most promising strategies in CRC therapy is the development of delivery systems based on nanomaterials that can transport antitumor agents to the tumor site more efficiently, increasing accumulation within the tumor and thus the antitumor effect. In addition to taking advantage of the increased permeability and retention effect (EPR) of solid tumors, these nanoformulations can be conjugated with monoclonal antibodies that recognize molecular markers that are specifically over-expressed on CRC cells. Active targeting of nanoformulations reduces the adverse effects associated with the cytotoxic activity of drugs in healthy tissues, which will be of interest for improving the quality of life of cancer patients in the future. This review focuses on in vitro and in vivo studies of drug delivery nanoformulations functionalized with monoclonal antibodies for targeted therapy of CRC.
Collapse
Affiliation(s)
- Ana Cepero
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Granada, 18014, Spain
| | - Cristina Luque
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Granada, 18014, Spain
| | - Laura Cabeza
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Granada, 18014, Spain
| | - Gloria Perazzoli
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain
| | - Francisco Quiñonero
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Granada, 18014, Spain
| | - Cristina Mesas
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Granada, 18014, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Granada, 18014, Spain,Correspondence: Consolación Melguizo, Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain, Tel +34-958-249833, Email
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, 18100, Spain,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18071, Spain,Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Granada, 18014, Spain
| |
Collapse
|
11
|
Sun Y, Li B, Cao Q, Liu T, Li J. Targeting cancer stem cells with polymer nanoparticles for gastrointestinal cancer treatment. Stem Cell Res Ther 2022; 13:489. [PMID: 36182897 PMCID: PMC9526954 DOI: 10.1186/s13287-022-03180-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 09/24/2022] [Indexed: 11/10/2022] Open
Abstract
Nanomaterials are developing rapidly in the medical field, bringing new hope for treating various refractory diseases. Among them, polymer nanomaterials, with their excellent properties, have been used to treat various diseases, such as malignant tumors, diabetes, and nervous system diseases. Gastrointestinal cancer is among the cancers with the highest morbidity and mortality worldwide. Cancer stem cells are believed to play an important role in the occurrence and development of tumors. This article summarizes the characteristics of gastrointestinal cancer stem cells and reviews the latest research progress in treating gastrointestinal malignant tumors using polymer nanoparticles to target cancer stem cells. In addition, the review article highlights the potential of polymer nanoparticles in targeting gastrointestinal cancer stem cells.
Collapse
Affiliation(s)
- Yao Sun
- Department of General Surgery, The Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun, 130041, China
| | - Bo Li
- Department of Rehabilitation Medicine, China-Japan Union Hospital of Jilin University, Changchun, 130000, China
| | - Qian Cao
- Department of Education, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Tongjun Liu
- Department of General Surgery, The Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun, 130041, China
| | - Jiannan Li
- Department of General Surgery, The Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun, 130041, China.
| |
Collapse
|
12
|
Munro MJ, Wickremesekera SK, Tan ST, Peng L. Proteomic analysis of low- and high-grade human colon adenocarcinoma tissues and tissue-derived primary cell lines reveals unique biological functions of tumours and new protein biomarker candidates. Clin Proteomics 2022; 19:27. [PMID: 35842572 PMCID: PMC9287856 DOI: 10.1186/s12014-022-09364-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/22/2022] [Indexed: 11/10/2022] Open
Abstract
Background Colon cancer is the third most common cancer and second highest cause of cancer deaths worldwide. The aim of the study was to find new biomarkers for diagnosis, prognosis and therapeutic drug targets for this disease. Methods Four low-grade and four high-grade human colon adenocarcinoma tumours with patient-matched normal colon tissues were analysed. Additionally, tissue-derived primary cell lines were established from each tumour tissue. The cell lines were validated using DNA sequencing to confirm that they are a suitable in vitro model for colon adenocarcinoma based on conserved gene mutations. Label-free quantitation proteomics was performed to compare the proteomes of colon adenocarcinoma samples to normal colon samples, and of colon adenocarcinoma tissues to tissue-derived cell lines to find significantly differentially abundant proteins. The functions enriched within the differentially expressed proteins were assessed using STRING. Proteomics data was validated by Western blotting. Results A total of 4767 proteins were identified across all tissues, and 4711 across primary tissue-derived cell lines. Of these, 3302 proteins were detected in both the tissues and the cell lines. On average, primary cell lines shared about 70% of proteins with their parent tissue, and they retained mutations to key colon adenocarcinoma-related genes and did not diverge far genetically from their parent tissues. Colon adenocarcinoma tissues displayed upregulation of RNA processing, steroid biosynthesis and detoxification, and downregulation of cytoskeletal organisation and loss of normal muscle function. Tissue-derived cell lines exhibited increased interferon-gamma signalling and aberrant ferroptosis. Overall, 318 proteins were significantly up-regulated and 362 proteins significantly down-regulated by comparisons of high-grade with low-grade tumours and low-grade tumour with normal colon tissues from both sample types. Conclusions The differences exhibited between tissues and cell lines highlight the additional information that can be obtained from patient-derived primary cell lines. DNA sequencing and proteomics confirmed that these cell lines can be considered suitable in vitro models of the parent tumours. Various potential biomarkers for colon adenocarcinoma initiation and progression and drug targets were identified and discussed, including seven novel markers: ACSL4, ANK2, AMER3, EXOSC1, EXOSC6, GCLM, and TFRC. Supplementary Information The online version contains supplementary material available at 10.1186/s12014-022-09364-y.
Collapse
Affiliation(s)
- Matthew J Munro
- School of Biological Sciences and Centre for Biodiscovery, Victoria University of Wellington, Wellington, 6140, New Zealand.,Gillies McIndoe Research Institute, Newtown, PO Box 7184, Wellington, 6242, New Zealand
| | - Susrutha K Wickremesekera
- Gillies McIndoe Research Institute, Newtown, PO Box 7184, Wellington, 6242, New Zealand.,Upper Gastrointestinal, Hepatobiliary & Pancreatic Section, Department of General Surgery, Wellington Regional Hospital, Wellington, 6021, New Zealand
| | - Swee T Tan
- Gillies McIndoe Research Institute, Newtown, PO Box 7184, Wellington, 6242, New Zealand. .,Wellington Regional Plastic, Maxillofacial & Burns Unit, Hutt Hospital, Lower Hutt, 5040, New Zealand. .,Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3050, Australia.
| | - Lifeng Peng
- School of Biological Sciences and Centre for Biodiscovery, Victoria University of Wellington, Wellington, 6140, New Zealand.
| |
Collapse
|
13
|
Emran TB, Shahriar A, Mahmud AR, Rahman T, Abir MH, Siddiquee MFR, Ahmed H, Rahman N, Nainu F, Wahyudin E, Mitra S, Dhama K, Habiballah MM, Haque S, Islam A, Hassan MM. Multidrug Resistance in Cancer: Understanding Molecular Mechanisms, Immunoprevention and Therapeutic Approaches. Front Oncol 2022; 12:891652. [PMID: 35814435 PMCID: PMC9262248 DOI: 10.3389/fonc.2022.891652] [Citation(s) in RCA: 222] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/10/2022] [Indexed: 12/15/2022] Open
Abstract
Cancer is one of the leading causes of death worldwide. Several treatments are available for cancer treatment, but many treatment methods are ineffective against multidrug-resistant cancer. Multidrug resistance (MDR) represents a major obstacle to effective therapeutic interventions against cancer. This review describes the known MDR mechanisms in cancer cells and discusses ongoing laboratory approaches and novel therapeutic strategies that aim to inhibit, circumvent, or reverse MDR development in various cancer types. In this review, we discuss both intrinsic and acquired drug resistance, in addition to highlighting hypoxia- and autophagy-mediated drug resistance mechanisms. Several factors, including individual genetic differences, such as mutations, altered epigenetics, enhanced drug efflux, cell death inhibition, and various other molecular and cellular mechanisms, are responsible for the development of resistance against anticancer agents. Drug resistance can also depend on cellular autophagic and hypoxic status. The expression of drug-resistant genes and the regulatory mechanisms that determine drug resistance are also discussed. Methods to circumvent MDR, including immunoprevention, the use of microparticles and nanomedicine might result in better strategies for fighting cancer.
Collapse
Affiliation(s)
- Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Asif Shahriar
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, United States
| | - Aar Rafi Mahmud
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
| | - Tanjilur Rahman
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Mehedy Hasan Abir
- Faculty of Food Science and Technology, Chattogram Veterinary and Animal Sciences University, Chattogram, Bangladesh
| | | | - Hossain Ahmed
- Department of Biotechnology and Genetic Engineering, University of Development Alternative, Dhaka, Bangladesh
| | - Nova Rahman
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Dhaka, Bangladesh
| | - Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Elly Wahyudin
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Mahmoud M Habiballah
- Medical Laboratory Technology Department, Jazan University, Jazan, Saudi Arabia
- SMIRES for Consultation in Specialized Medical Laboratories, Jazan University, Jazan, Saudi Arabia
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
- Bursa Uludağ University Faculty of Medicine, Bursa, Turkey
| | | | - Mohammad Mahmudul Hassan
- Queensland Alliance for One Health Sciences, School of Veterinary Science, The University of Queensland, Gatton, QLD, Australia
- Department of Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University, Chattogram, Bangladesh
| |
Collapse
|
14
|
Nie H, Wu Y, Ou C, He X. Comprehensive Analysis of SMC Gene Family Prognostic Value and Immune Infiltration in Patients With Pancreatic Adenocarcinoma. Front Med (Lausanne) 2022; 9:832312. [PMID: 35372377 PMCID: PMC8965256 DOI: 10.3389/fmed.2022.832312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/09/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic adenocarcinoma (PAAD) is a malignant tumor with high morbidity and mortality worldwide. Members from the structural maintenance of chromosomes (SMC) gene family function as oncogenes in various tumor types, but their roles in PAAD have not been elucidated. In this study, we aimed to explore the role of the SMC family in tumor progression and cancer immune infiltration in PAAD using integrative bioinformatic analyses. The results showed that the SMC 1A, 2, 3, 4, and 6 were overexpressed in PAAD tissues; of these, SMC 1A, 4, 5, and 6 could be potential prognostic biomarkers for PAAD. The expression of SMC genes was found to be strongly associated with immune cell infiltration. According to the infiltrative status of various immune cells, the mRNA expression of SMC genes in PAAD was associated with the overall and recurrence-free survival of patients. In conclusion, the SMC gene family is associated with PAAD and may be involved in tumorigenesis and cancer-immune interactions; thus, members from this gene family may serve as promising prognostic and therapeutic biomarkers of PAAD.
Collapse
Affiliation(s)
- Hui Nie
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Yanhao Wu
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyun He
- Departments of Ultrasound Imaging, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
15
|
Marschall ALJ. Targeting the Inside of Cells with Biologicals: Chemicals as a Delivery Strategy. BioDrugs 2021; 35:643-671. [PMID: 34705260 PMCID: PMC8548996 DOI: 10.1007/s40259-021-00500-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2021] [Indexed: 12/17/2022]
Abstract
Delivering macromolecules into the cytosol or nucleus is possible in vitro for DNA, RNA and proteins, but translation for clinical use has been limited. Therapeutic delivery of macromolecules into cells requires overcoming substantially higher barriers compared to the use of small molecule drugs or proteins in the extracellular space. Breakthroughs like DNA delivery for approved gene therapies and RNA delivery for silencing of genes (patisiran, ONPATTRO®, Alnylam Pharmaceuticals, Cambridge, MA, USA) or for vaccination such as the RNA-based coronavirus disease 2019 (COVID-19) vaccines demonstrated the feasibility of using macromolecules inside cells for therapy. Chemical carriers are part of the reason why these novel RNA-based therapeutics possess sufficient efficacy for their clinical application. A clear advantage of synthetic chemicals as carriers for macromolecule delivery is their favourable properties with respect to production and storage compared to more bioinspired vehicles like viral vectors or more complex drugs like cellular therapies. If biologicals can be applied to intracellular targets, the druggable space is substantially broadened by circumventing the limited utility of small molecules for blocking protein–protein interactions and the limitation of protein-based drugs to the extracellular space. An in depth understanding of the macromolecular cargo types, carrier types and the cell biology of delivery is crucial for optimal application and further development of biologicals inside cells. Basic mechanistic principles of the molecular and cell biological aspects of cytosolic/nuclear delivery of macromolecules, with particular consideration of protein delivery, are reviewed here. The efficiency of macromolecule delivery and applications in research and therapy are highlighted.
Collapse
Affiliation(s)
- Andrea L J Marschall
- Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Brunswick, Germany.
| |
Collapse
|
16
|
Wang H, Chen Y, Yang D, Ma L. Perspective of Human Condensins Involved in Colorectal Cancer. Front Pharmacol 2021; 12:664982. [PMID: 34557090 PMCID: PMC8453263 DOI: 10.3389/fphar.2021.664982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 08/19/2021] [Indexed: 12/24/2022] Open
Abstract
Although many important roles are played by human condesins in condensation and segregation of mitotic chromosomes, what roles of human condensins play in colorectal cancer are still unclear at present. Recently, abnormal expressions of all eight subunits of human condensins have been found in colorectal cancer and they are expected to become potential biomarkers and therapeutic targets for colorectal cancer in the future. However, there are still no reviews on the significance of abnormal expression of human condensin subunits and colorectal cancer until now. Based on a brief introduction to the discovery and composition of human condensins, the review summarized all abnormally expressed human subunits found in colorectal cancer based on publicly published papers. Moreover, Perspective of application on abnormally expressed human subunits in colorectal cancer is further reviewed.
Collapse
Affiliation(s)
- Hongzhen Wang
- School of Life Sciences, Jilin Normal University, Siping, China
| | - Yao Chen
- School of Life Sciences, Jilin Normal University, Siping, China
| | - Dawei Yang
- The Department of General Surgery, The Central People's Hospital of Siping City, Siping, China
| | - Liang Ma
- The Department of General Surgery, The Central People's Hospital of Siping City, Siping, China
| |
Collapse
|
17
|
Oliveira BSAD, de Assis ACC, Souza NM, Ferreira LFR, Soriano RN, Bilal M, Iqbal HMN. Nanotherapeutic approach to tackle chemotherapeutic resistance of cancer stem cells. Life Sci 2021; 279:119667. [PMID: 34087280 DOI: 10.1016/j.lfs.2021.119667] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/16/2021] [Accepted: 05/22/2021] [Indexed: 02/07/2023]
Abstract
Estimates indicate that cancer will become the leading cause of mortality worldwide in the future. Tumorigenesis is a complex process that involves self-sufficiency in signs of growth, insensitivity to anti-growth signals, prevention of apoptosis, unlimited replication, sustained angiogenesis, tissue invasion, and metastasis. Cancer stem cells (CSCs) have an important role in tumor development and resistance. Here we will approach phenotypic plasticity capacity, highly efficient DNA repair systems, anti-apoptotic machinery, sustained stemness features, interaction with the tumor microenvironment, and Notch, Wnt, and Hedgehog signaling pathways. The researches about CSCs as a target in cancer treatment has been growing. Many different options have pointed beneficial results, such as pathways and CSC-surface markers targeting. Besides its limitations, nanotherapeutics have emerged as a potential strategy in this context since they aim to improve pharmacokinetics, biodistribution, and reduce the side effects observed in traditional treatments. Nanoparticles have been studied in this field, mostly for drug delivery and a multitherapy approach. Another widely researched approaches in this area are related to heat therapy, such as photothermal therapy, photodynamic therapy and magnetic hyperthermia, besides molecular targeting. This review will contemplate the most relevant studies that have shown the effects of nanotherapeutics. In conclusion, although the studies analyzed are mostly preclinical, we believe that there is strong evidence that nanoparticles can increase the chances of a better prognosis to cancer in the future. It is also essential to transpose these findings to the clinic to confirm and better understand the role of nanotherapeutics in this context.
Collapse
Affiliation(s)
- Bruna Stefane Alves de Oliveira
- Undergradute student, Department of Medicine, Federal University of Juiz de Fora, Governador Valadares, MG 35032-620, Brazil
| | - Ana Carolina Correa de Assis
- Undergradute student, Department of Medicine, Federal University of Juiz de Fora, Governador Valadares, MG 35032-620, Brazil
| | - Natália Melo Souza
- Undergradute student, Department of Medicine, Federal University of Juiz de Fora, Governador Valadares, MG 35032-620, Brazil
| | - Luiz Fernando Romanholo Ferreira
- Graduate Program in Process Engineering, Tiradentes University (UNIT), Av. Murilo Dantas, 300, Farolândia, 49032-490 Aracaju, Sergipe, Brazil; Institute of Technology and Research (ITP), Tiradentes University (UNIT), Av. Murilo Dantas, 300, Farolândia, 49032-490 Aracaju, Sergipe, Brazil
| | - Renato Nery Soriano
- Division of Physiology and Biophysics, Department of Basic Life Sciences, Federal University of Juiz de Fora, Governador Valadares, MG 35010-177, Brazil
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian 223003, China.
| | - Hafiz M N Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico.
| |
Collapse
|
18
|
Jiang L, Guo T, Jiang Y, Liu P, Bai Y. Dauricine inhibits human pancreatic carcinoma cell proliferation through regulating miRNAs. Mol Omics 2021; 17:630-640. [PMID: 34184018 DOI: 10.1039/d1mo00156f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Pancreatic cancer is one of the most malignant digestive tract tumors with the worst prognosis. Dauricine (Dau) can inhibit the proliferation of the pancreatic cancer cell line, and has the potential to be used as an adjuvant drug against pancreatic cancer; however, the working mechanism of Dau has not been elucidated. To unravel the effects and mechanisms of Dau on proteins and metabolic pathways, we evaluated the mRNA and microRNA expression in BxPC3 cells treated with Dau. The differences in the gene expression were compared using principal component analysis using mRNA and miRNA data to detect and analyze the sample discrimination. 187 miRNA and 907 mRNA that were significantly differentially expressed were identified using Python programming. On comparing genes and miRNAs in the DISEASES database, 79 known miRNA and 47 mRNA were found to be affected by Dau. The up-regulated and down-regulated genes were annotated with GO biological processes to determine the functional effect. Interactions between mRNA and mRNA were analyzed using the STRING database and the miRBase database was queried to obtain experimentally verified interactions between miRNA and mRNA as edges of miRNA and mRNA in the network. Finally, 413 sites and 2125 sides of the network were obtained, including 1 up-regulated and 18 down-regulated miRNAs. The expression of 19 miRNAs was identified by qPCR. The analysis of the protein-protein interaction network, using the Molecular Complex Detection (MCODE) plug-in of cytoscape, helped in identifying 12 important sub-networks. Most subnets are indirectly or directly related to specific miRNAs. This study provides evidence for the anticancer effect of Dau as a potential anticancer compound.
Collapse
Affiliation(s)
- Ling Jiang
- School Basic Medical Science, Heilongjiang University of Chinese Medicine, Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, 150040, P. R. China.
| | - Tianzhu Guo
- School Basic Medical Science, Heilongjiang University of Chinese Medicine, Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, 150040, P. R. China.
| | - Ying Jiang
- School Basic Medical Science, Heilongjiang University of Chinese Medicine, Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, 150040, P. R. China.
| | - Ping Liu
- School Basic Medical Science, Heilongjiang University of Chinese Medicine, Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, 150040, P. R. China.
| | - Yun Bai
- School Basic Medical Science, Heilongjiang University of Chinese Medicine, Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, 150040, P. R. China.
| |
Collapse
|
19
|
Esmaeili SA, Sahranavard S, Salehi A, Bagheri V. Selectively targeting cancer stem cells: Current and novel therapeutic strategies and approaches in the effective eradication of cancer. IUBMB Life 2021; 73:1045-1059. [PMID: 34184810 DOI: 10.1002/iub.2524] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/19/2022]
Abstract
Cancer stem cells (CSCs) are a subgroup of cells in malignant cancers, which possess self-renewal capacity, tumor-initiating capability, and pluripotency, as well as being responsible for tumor maintenance, metastasis, relapse, and chemoresistance. The treatment modalities previously established for cancer included surgery, chemotherapy, and radiotherapy. The majority of tumor cells of non-CSCs could be eradicated using conventional chemotherapy and radiotherapy. Therefore, novel and promising therapeutic strategies that selectively target CSCs are of great importance. In this review, we described different therapeutic strategies such as immunotherapy, metabolism-based therapeutic strategies, and additional potential therapeutic approaches (targeting microRNAs [miRNAs], histone deacetylase, and DNA methyl transferase) against CSCs. Taken together, due to the inefficiency of anticancer single therapies, targeting CSCs through their metabolism and using immunotherapy and miRNAs besides classical chemo- and radiotherapy may exert better therapeutic effects.
Collapse
Affiliation(s)
- Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shamim Sahranavard
- Department of Traditional Pharmacy, School of Traditional Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Astireh Salehi
- Biology Department, Islamic Azad University, Sanandaj, Iran
| | - Vahid Bagheri
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
20
|
Ortíz R, Quiñonero F, García-Pinel B, Fuel M, Mesas C, Cabeza L, Melguizo C, Prados J. Nanomedicine to Overcome Multidrug Resistance Mechanisms in Colon and Pancreatic Cancer: Recent Progress. Cancers (Basel) 2021; 13:2058. [PMID: 33923200 PMCID: PMC8123136 DOI: 10.3390/cancers13092058] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/22/2021] [Accepted: 04/22/2021] [Indexed: 12/24/2022] Open
Abstract
The development of drug resistance is one of the main causes of cancer treatment failure. This phenomenon occurs very frequently in different types of cancer, including colon and pancreatic cancers. However, the underlying molecular mechanisms are not fully understood. In recent years, nanomedicine has improved the delivery and efficacy of drugs, and has decreased their side effects. In addition, it has allowed to design drugs capable of avoiding certain resistance mechanisms of tumors. In this article, we review the main resistance mechanisms in colon and pancreatic cancers, along with the most relevant strategies offered by nanodrugs to overcome this obstacle. These strategies include the inhibition of efflux pumps, the use of specific targets, the development of nanomedicines affecting the environment of cancer-specific tissues, the modulation of DNA repair mechanisms or RNA (miRNA), and specific approaches to damage cancer stem cells, among others. This review aims to illustrate how advanced nanoformulations, including polymeric conjugates, micelles, dendrimers, liposomes, metallic and carbon-based nanoparticles, are allowing to overcome one of the main limitations in the treatment of colon and pancreatic cancers. The future development of nanomedicine opens new horizons for cancer treatment.
Collapse
Affiliation(s)
- Raúl Ortíz
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (R.O.); (F.Q.); (B.G.-P.); (M.F.); (C.M.); (L.C.); (J.P.)
- Department of Anatomy and Embriology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Francisco Quiñonero
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (R.O.); (F.Q.); (B.G.-P.); (M.F.); (C.M.); (L.C.); (J.P.)
- Department of Anatomy and Embriology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Beatriz García-Pinel
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (R.O.); (F.Q.); (B.G.-P.); (M.F.); (C.M.); (L.C.); (J.P.)
- Department of Anatomy and Embriology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Marco Fuel
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (R.O.); (F.Q.); (B.G.-P.); (M.F.); (C.M.); (L.C.); (J.P.)
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Cristina Mesas
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (R.O.); (F.Q.); (B.G.-P.); (M.F.); (C.M.); (L.C.); (J.P.)
- Department of Anatomy and Embriology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Laura Cabeza
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (R.O.); (F.Q.); (B.G.-P.); (M.F.); (C.M.); (L.C.); (J.P.)
- Department of Anatomy and Embriology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (R.O.); (F.Q.); (B.G.-P.); (M.F.); (C.M.); (L.C.); (J.P.)
- Department of Anatomy and Embriology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (R.O.); (F.Q.); (B.G.-P.); (M.F.); (C.M.); (L.C.); (J.P.)
- Department of Anatomy and Embriology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| |
Collapse
|
21
|
Lv W, Champion JA. Demonstration of intracellular trafficking, cytosolic bioavailability, and target manipulation of an antibody delivery platform. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 32:102315. [PMID: 33065253 DOI: 10.1016/j.nano.2020.102315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/30/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023]
Abstract
Intracellular antibody delivery into live cells has significant implications for research and therapeutic applications. However, many delivery systems lack potency due to low uptake and/or endosomal entrapment and understanding of intracellular delivery processes is lacking. Herein, we studied the cellular uptake, intracellular trafficking and targeting of antibodies using our previously developed Hex antibody nanocarrier. We demonstrated Hex-antibodies were internalized through multiple endocytic routes into lysosomes and provide evidence of endo/lysosomal disruption and Hex-antibody release to the cytosol. Cytosolic antibodies retained their bioactivity for at least 24 h. Functional effect of Hex delivered anti-STAT3 antibodies was evidenced by inhibition of nuclear translocation of cytosolic transcription factor STAT3. This study has generated understanding of key steps in the Hex intracellular antibody delivery system and will facilitate the development of effective cytosolic antibody delivery and applications in both the therapeutic and research domains.
Collapse
Affiliation(s)
- Wei Lv
- School of Chemical & Biomolecular Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States
| | - Julie A Champion
- School of Chemical & Biomolecular Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States.
| |
Collapse
|