1
|
Tavabie OD, Salehi S, Aluvihare VR. The challenges and potential of microRNA-based therapy for patients with liver failure syndromes and hepatocellular carcinoma. Expert Opin Ther Targets 2024; 28:179-191. [PMID: 38487923 DOI: 10.1080/14728222.2024.2331598] [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: 12/21/2023] [Accepted: 03/13/2024] [Indexed: 03/20/2024]
Abstract
INTRODUCTION Morbidity and mortality from liver disease continues to rise worldwide. There are currently limited curative treatments for patients with liver failure syndromes, encompassing acute liver failure and decompensated cirrhosis states, outside of transplantation. Whilst there have been improvements in therapeutic options for patients with hepatocellular carcinoma (HCC), there remain challenges necessitating novel therapeutic agents. microRNA have long been seen as potential therapeutic targets but there has been limited clinical translation. AREAS COVERED We will discuss the limitations of conventional non-transplant management of patients with liver failure syndromes and HCC. We will provide an overview of microRNA and the challenges in developing and delivering microRNA-based therapeutic agents. We will finally provide an overview of microRNA-based therapeutic agents which have progressed to clinical trials. EXPERT OPINION microRNA have great potential to be developed into therapeutic agents due to their association with critical biological processes which govern health and disease. Utilizing microRNA sponges to target multiple microRNA associated with specific biological processes may improve their therapeutic efficacy. However, there needs to be significant improvements in delivery systems to ensure the safe delivery of microRNA to target sites and minimize systemic distribution. This currently significantly impacts the clinical translation of microRNA-based therapeutic agents.
Collapse
Affiliation(s)
| | - Siamak Salehi
- Institute of Liver Studies, King's College Hospital, London, UK
| | | |
Collapse
|
2
|
Badami E, Carcione C, Chinnici CM, Tinnirello R, Conaldi PG, Iannolo G. HCV Interplay With Mir34a: Implications in Hepatocellular Carcinoma. Front Oncol 2022; 11:803278. [PMID: 35127513 PMCID: PMC8812294 DOI: 10.3389/fonc.2021.803278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/27/2021] [Indexed: 12/24/2022] Open
Abstract
Since its identification, HCV has been considered one of the main causes of hepatitis and liver cancer. Currently, the molecular mechanisms of HCC development induced by HCV infection have not been sufficiently clarified. The recent discovery of novel treatments that inhibit HCV replication gave rise to new questions concerning HCC mechanisms. In particular, the HCV eradication mediated by new direct-acting antiviral (DAAs) drugs does not exclude the possibility of de novo HCC development; this finding opened more questions on the interplay between liver cells and the virus. Different groups have investigated the pathways leading to cancer recurrence in patients treated with DAAs. For this reason, we tried to gain molecular insights into the changes induced by HCV infection in the target liver cells. In particular, we observed an increase in microRNA34a (miR34a) expression following HCV infection of HCC cell line Huh7.5. In addition, Huh7.5 treated with extracellular vesicles (EVs) from the previously HCV-infected Huh7.5 underwent apoptosis. Since miR34 expression was increased in Huh7.5 EVs, we hypothesized a paracrine mechanism of viral infection mediated by miR34a cargo of EVs. The balance between viral infection and cell transformation may raise some questions on the possible use of antiviral drugs in association with antineoplastic treatment.
Collapse
Affiliation(s)
- Ester Badami
- Department of Research, Istituto di Ricovero e Cura a Carattere Scientifico Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (IRCCS ISMETT), Palermo, Italy
- Regenerative Medicine and Immunotherapy Area, Fondazione Ri.MED, Palermo, Italy
| | - Claudia Carcione
- Regenerative Medicine and Immunotherapy Area, Fondazione Ri.MED, Palermo, Italy
| | - Cinzia Maria Chinnici
- Department of Research, Istituto di Ricovero e Cura a Carattere Scientifico Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (IRCCS ISMETT), Palermo, Italy
- Regenerative Medicine and Immunotherapy Area, Fondazione Ri.MED, Palermo, Italy
| | - Rosaria Tinnirello
- Neuroscience Unit, Consiglio Nazionale delle Ricerche (CNR), Institute of Biomedicine and Molecular Immunology, Palermo, Italy
| | - Pier Giulio Conaldi
- Department of Research, Istituto di Ricovero e Cura a Carattere Scientifico Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (IRCCS ISMETT), Palermo, Italy
| | - Gioacchin Iannolo
- Department of Research, Istituto di Ricovero e Cura a Carattere Scientifico Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (IRCCS ISMETT), Palermo, Italy
- *Correspondence: Gioacchin Iannolo, ; ; orcid.org/0000-0002-7710-4735
| |
Collapse
|
3
|
Hong DS, Kang YK, Borad M, Sachdev J, Ejadi S, Lim HY, Brenner AJ, Park K, Lee JL, Kim TY, Shin S, Becerra CR, Falchook G, Stoudemire J, Martin D, Kelnar K, Peltier H, Bonato V, Bader AG, Smith S, Kim S, O'Neill V, Beg MS. Phase 1 study of MRX34, a liposomal miR-34a mimic, in patients with advanced solid tumours. Br J Cancer 2020; 122:1630-1637. [PMID: 32238921 PMCID: PMC7251107 DOI: 10.1038/s41416-020-0802-1] [Citation(s) in RCA: 476] [Impact Index Per Article: 119.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 08/14/2019] [Accepted: 03/04/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND In this first-in-human, Phase 1 study of a microRNA-based cancer therapy, the recommended Phase 2 dose (RP2D) of MRX34, a liposomal mimic of microRNA-34a (miR-34a), was determined and evaluated in patients with advanced solid tumours. METHODS Adults with various solid tumours refractory to standard treatments were enrolled in 3 + 3 dose-escalation cohorts and, following RP2D determination, expansion cohorts. MRX34, with oral dexamethasone premedication, was given intravenously daily for 5 days in 3-week cycles. RESULTS Common all-cause adverse events observed in 85 patients enrolled included fever (% all grade/G3: 72/4), chills (53/14), fatigue (51/9), back/neck pain (36/5), nausea (36/1) and dyspnoea (25/4). The RP2D was 70 mg/m2 for hepatocellular carcinoma (HCC) and 93 mg/m2 for non-HCC cancers. Pharmacodynamic results showed delivery of miR-34a to tumours, and dose-dependent modulation of target gene expression in white blood cells. Three patients had PRs and 16 had SD lasting ≥4 cycles (median, 19 weeks, range, 11-55). CONCLUSION MRX34 treatment with dexamethasone premedication demonstrated a manageable toxicity profile in most patients and some clinical activity. Although the trial was closed early due to serious immune-mediated AEs that resulted in four patient deaths, dose-dependent modulation of relevant target genes provides proof-of-concept for miRNA-based cancer therapy. CLINICAL TRIAL REGISTRATION NCT01829971.
Collapse
Grants
- P30 CA016672 NCI NIH HHS
- Research/Grant Funding: AbbVie, Adaptimmune, Amgen, Astra-Zeneca, Bayer, BMS, Daiichi-Sankyo, Eisai, Fate Therapeutics, Genentech, Genmab, Ignyta, Infinity, Kite, Kyowa, Lilly, LOXO, Merck, MedImmune, Mirati, MiRNA, Molecular Templates, Mologen, NCI-CTEP, Novartis, Pfizer, Seattle Genetics, Takeda; Travel, Accommodations, Expenses: LOXO, MiRNA; Consulting or Advisory Role: Alpha Insights, Axiom, Adaptimmune, Baxter, Bayer (Ad Board and Speakers Bureau), Genentech, GLG, Group H, Guidepoint Global, Infinity, Janssen, Merrimack, Medscape, Numab, Pfizer, Seattle Genetics, Takeda, Trieza Therapeutics Other ownership interests: Molecular Match (Advisor), OncoResponse (founder), Presagia Inc (Advisor)
- Consulting or Advisory Role: Lilly/ImClone; Novartis; Ono Pharmaceutical; Roche/ Genentech; Taiho Pharmaceutical; Research Funding: Bayer; Novartis; Roche/Genentech
- Honoraria: Celgene; Consulting or Advisory Role: Celgene
- Honoraria: Vascular Biogenics; Consulting or Advisory Role: NanoTX; Teleflex Medical Research Funding: Mirna Therapeutics (Inst); Threshold Pharmaceuticals; Patents, Royalties, Other Intellectual Property: NanoTx Pharmaceuticals; Travel, Accommodations, Expenses: Vascular Biogenics
- Royalties: Wolters Kluwer; Advisory role: EMD Serono; Travel: Bristol-Myers Squibb, EMD Serono, Millennium; Research funding: 3-V Biosciences, Abbvie, Aileron, American Society of Clinical Oncology, Amgen, ARMO, AstraZeneca, BeiGene, Biothera, Celldex, Celgene, Ciclomed, Curegenix, Curis, DelMar, eFFECTOR, Eli Lilly, EMD Serono, Fujifilm, Genmab, GlaxoSmithKline, Hutchison MediPharma, Ignyta, Incyte, Jacobio, Jounce, Kolltan, Loxo, MedImmune, Millennium, Merck, miRNA Therapeutics, National Institutes of Health, Novartis, OncoMed, Oncothyreon, Precision Oncology, Regeneron, Rgenix, Strategia, Syndax, Taiho, Takeda, Tarveda, Tesaro, Tocagen, U.T. MD Anderson Cancer Center, Vegenics
- Employment: Mirna Therapeutics; Stock and Other Ownership Interests: Mirna Therapeutics
- Employment: Mirna Therapeutics; Leadership: Mirna Therapeutics; Stock and Other Ownership Interests: Mirna Therapeutics; Pfizer; Patents, Royalties, Other Intellectual Property: Listed as an inventor on patent applications, but no ownership interest or royalties.
Collapse
Affiliation(s)
- David S Hong
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | | | | | - Jasgit Sachdev
- Scottsdale Healthcare Research Institute, Scottsdale, AZ, USA
| | - Samuel Ejadi
- University of California Irvine Medical Center, Orange, CA, USA
| | | | - Andrew J Brenner
- The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | | | | | - Tae-You Kim
- Seoul National University Hospital, Seoul, South Korea
| | | | - Carlos R Becerra
- Texas Oncology-US Oncology-Baylor University Medical Center, Dallas, TX, USA
| | - Gerald Falchook
- Sarah Cannon Research Institute at HealthONE, Denver, CO, USA
| | | | | | | | | | | | | | | | | | | | - Muhammad S Beg
- The University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
4
|
Bai J, Wu J, Tang R, Sun C, Ji J, Yin Z, Ma G, Yang W. Emodin, a natural anthraquinone, suppresses liver cancer in vitro and in vivo by regulating VEGFR 2 and miR-34a. Invest New Drugs 2019; 38:229-245. [PMID: 30976957 DOI: 10.1007/s10637-019-00777-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 04/02/2019] [Indexed: 01/02/2023]
Abstract
The pharmacokinetic (PK) and potential effects of Emodin on liver cancer were systematically evaluated in this study. Both the intragastric administration (i.g.) and hypodermic injection (i.h.) of Emodin exhibited a strong absorption (absorption rate < 1 h) and elimination capacity (t1/2 ≈ 2 h). The tissue distribution of Emodin after i.h. was rapid and wide. The stability of Emodin in three species of liver microsomes wasrat >human> beagle dog. These PK data provided the basis for the subsequent animal experiments. In liver cancer patient tissues, the expression of vascular endothelial growth factor (VEGF)-induced signaling pathways, including phosphorylated VEGF receptor 2 (VEGFR2), AKT, and ERK1/2,were simultaneously elevated, but miR-34a expression was reduced and negatively correlated with SMAD2 and SMAD4. Emodin inhibited the expression of SMAD2/4 in HepG2 cells by inducing the miR-34a level. Subsequently, BALB/c nude mice received a daily subcutaneous injection of HepG2 cells with or without Emodin treatment (1 mg/kg or 10 mg/kg), and Emodin inhibited tumorigenesis and reduced the mortality rate in a dose-dependent manner. In vivo experiments showed that cell proliferation, migration, and invasion were promoted by VEGF or miR-34a signal treatment but were inhibited when combined with Emodin treatment. All these results demonstrated that Emodin inhibited tumorigenesis in liver cancer by simultaneously inhibiting the VEGFR2-AKT-ERK1/2signaling pathway and promoting a miR-34a-mediated signaling pathway.
Collapse
Affiliation(s)
- Jianguo Bai
- Department of Hepatobiliary Surgery, the Fourth Hospital of Hebei Medical University, NO.12, Jiankang Road, Shijiazhuang, 050011, Hebei Province, People's Republic of China
| | - Jianfei Wu
- Department of Hepatobiliary Surgery, the Affiliated Hospital of Hebei University, Baoding, 071000, Hebei Province, People's Republic of China
| | - Ruifeng Tang
- Department of Hepatobiliary Surgery, the Fourth Hospital of Hebei Medical University, NO.12, Jiankang Road, Shijiazhuang, 050011, Hebei Province, People's Republic of China.
| | - Chao Sun
- Department of Hepatobiliary Surgery, the Fourth Hospital of Hebei Medical University, NO.12, Jiankang Road, Shijiazhuang, 050011, Hebei Province, People's Republic of China
| | - Junwei Ji
- Department of Emergency, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei Province, People's Republic of China
| | - Zhaolin Yin
- Department of ultrasound, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei Province, People's Republic of China
| | - Guangjun Ma
- Department of Hepatobiliary Surgery, the Fourth Hospital of Hebei Medical University, NO.12, Jiankang Road, Shijiazhuang, 050011, Hebei Province, People's Republic of China
| | - Wei Yang
- Department of Hepatobiliary Surgery, the Fourth Hospital of Hebei Medical University, NO.12, Jiankang Road, Shijiazhuang, 050011, Hebei Province, People's Republic of China
| |
Collapse
|
5
|
Li Y, Li C, Li D, Yang L, Jin J, Zhang B. lncRNA KCNQ1OT1 enhances the chemoresistance of oxaliplatin in colon cancer by targeting the miR-34a/ATG4B pathway. Onco Targets Ther 2019; 12:2649-2660. [PMID: 31040703 PMCID: PMC6462170 DOI: 10.2147/ott.s188054] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Purpose The chemoresistance of colon cancer to oxaliplatin (L-OHP) indicates poor prognosis. Long non-coding RNA (lncRNA) KCNQ1OT1 (KCNQ1 opposite strand/antisense transcript 1) has been shown to participate in the tumorigenesis of several types of cancers. However, little is known about the role of KCNQ1OT1 in the chemoresistance and prognosis of colon cancer. Materials and methods Quantitative-PCR and Western blot were used to measure the expression profiles of KCNQ1OT1, miR-34a, and Atg4B in colon cancer tissues and cells. Cell viability assay and flow cytometry were used to examine their effects on cell proliferation and death. Cleavage of LC3 and GFP-LC3 plasmid transfection were used to detect autophagic activity. Double luciferase reporter assay was used to verify the interactions between miRNA and lncRNA or mRNA. Xenograft tumor model was used to verify the effects of KCNQ1OT1 in vivo. Results In this study, it is shown that the expression level of KCNQ1OT1 was increased in tumor, which indicated poor prognosis in colon cancer patients. Using colon cancer cell lines HCT116 and SW480, it was demonstrated that knockdown of KCNQ1OT1 decreased the cell viability and increased the apoptosis rates upon L-OHP treatment. Further studies indicated that Atg4B upregulation was partially responsible for KCNQ1OT1-induced protective autophagy and chemoresistance. Moreover, miR-34a functioned as a bridge between KCNQ1OT1 and Atg4B, which could be sponged by KCNQ1OT1, while it could also bind to the 3'-UTR of Atg4B and downregulate its expressions. Finally, we show that the KCNQ1OT1/miR-34a/Atg4B axis regulated the chemoresistance of colon cancer cells in vitro and in vivo. Conclusion lncRNA KCNQ1OT1 promoted the chemoresistance of colon cancer by sponging miR-34a, thus upregulating the expressions of Atg4B and enhancing protective autophagy. KCNQ1OT1 might become a promising target for colon cancer therapeutics.
Collapse
Affiliation(s)
- Yongchao Li
- Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China,
| | - Dandan Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China,
| | - Lei Yang
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China,
| | - Jingpeng Jin
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China,
| | - Bin Zhang
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China,
| |
Collapse
|
6
|
Prognostic Value of MicroRNA-15a in Human Cancers: A Meta-Analysis and Bioinformatics. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2063823. [PMID: 31061821 PMCID: PMC6466945 DOI: 10.1155/2019/2063823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 02/22/2019] [Accepted: 03/05/2019] [Indexed: 02/06/2023]
Abstract
Background Although several studies have proved the relationship between the prognostic value of miRNA-15a and different types of cancer, the result remains controversial. Thus, a meta-analysis was conducted to clarify the prognostic value of miRNA-15a expression level in human cancers. Methods We enrolled appropriate literature by searching the databases of PubMed, Embase, and Web of Science. Subsequently, we extracted HRs and their 95% CIs and calculated pooled results of miRNA-15a for overall survival (OS) and disease-free survival (DFS). Besides, subgroup analysis, sensitivity analysis, and publication bias were also revealed in this study. We also further validated this meta-analysis using the Kaplan-Meier plotter database. Result 10 studies, including 1616 patients, were embraced in our meta-analysis. The result showed the lower expression of miRNA-15a significantly predicted adverse OS (HR=2.17, 95% CI: 1.41-3.34), but there is no significant association between the expressing level and DFS in cancer patient (HR=2.04, 95% CI: 0.60-6.88). Based on Kaplan-Meier plotter database, we found the same results in bladder Carcinoma, head-neck squamous cell carcinoma, liver hepatocellular carcinoma, lung squamous cell carcinoma, pancreatic ductal adenocarcinoma, rectum adenocarcinoma, stomach adenocarcinoma, and uterine corpus endometrial carcinoma, but opposite results were found in cervical squamous cell carcinoma and esophageal carcinoma. Conclusion Low expressing levels of miRNA-15a indicated poor OS, while miRNA-15a can be used as a prediction biomarker in different cancer types.
Collapse
|
7
|
Clinical Impact of Epithelial-to-Mesenchymal Transition Regulating MicroRNAs in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2018; 10:cancers10090328. [PMID: 30217058 PMCID: PMC6162771 DOI: 10.3390/cancers10090328] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 08/30/2018] [Accepted: 09/11/2018] [Indexed: 12/20/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive carcinoma entities worldwide with early and rapid dissemination. Recently, we discussed the role of microRNAs as epigenetic regulators of Epithelial-to-Mesenchymal Transition (EMT) in PDAC. In this study, we investigated their value as diagnostic and prognostic markers in tissue and blood samples of 185 patients including PDAC, non-malignant pancreatic disorders, and age-matched healthy controls. Expression of the microRNA-200-family (microRNAs -141, -200a, -200b, -200c, -429) and microRNA-148a was significantly downregulated in tissue of PDAC Union internationale contre le cancer (UICC) Stage II. Correspondingly, stromal PDAC tissue showed strong expression of Fibronectin, Vimentin, and ZEB-1 (Zinc finger E-box-binding homeobox) versus low expression of E-cadherin. Transient transfection of microRNA-200b and microRNA-200c mimics resulted in the downregulation of their key target ZEB-1. Inversely, blood serum analyses of patients with PDAC UICC Stages II, III, and IV showed a significant over-expression of microRNA-200-family members, microRNA-148a, microRNA-10b, and microRNA-34a. Correspondingly, Enzyme-linked Immunosorbent Assay (ELISA) analyses revealed a significant over-expression of soluble E-cadherin in serum samples of PDAC patients versus healthy controls. The best diagnostic accuracy to distinguish between PDAC and non-PDAC in this patient collective could be achieved in tissue by microRNA-148a with an area under the receiver-operating-characteristic (ROC) curve (AUC) of 0.885 and in blood serum by a panel of microRNA-141, -200b, -200c, and CA.19-9 with an AUC of 0.890. Both diagnostic tools outreach the diagnostic performance of the currently most common diagnostic biomarker CA.19-9 (AUC of 0.834). Kaplan Meier survival analysis of this patient collective revealed an improved overall survival in PDAC patients with high expression of tissue-related microRNA-34a, -141, -200b, -200c, and -429. In conclusion, EMT-regulating microRNAs have great potential as liquid and solid biopsy markers in PDAC patients. Their prognostic and therapeutic benefits remain important tasks for future studies.
Collapse
|
8
|
Sun H, Shao Y, Huang J, Sun S, Liu Y, Zhou P, Yang H. Prognostic value of microRNA-9 in cancers: a systematic review and meta-analysis. Oncotarget 2018; 7:67020-67032. [PMID: 27563807 PMCID: PMC5341854 DOI: 10.18632/oncotarget.11466] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 08/08/2016] [Indexed: 12/28/2022] Open
Abstract
Recent studies revealed that different microRNA-9 (miR-9) expressions were associated with prognoses of different cancers. We conducted this meta-analysis to evaluate the prognostic value of miR-9. PubMed, Embase, Web of Science, and Cochrane Library (last update by November 30, 2015) were searched for literatures. A total of 17 studies from 16 articles were finally qualified and enrolled in this meta-analysis. Pooled analyses showed that a higher expression of miR-9 might predict poor overall survival (HR: 2.17, 95% CI: 1.39 – 3.41, P < 0.001 (7.23 * 10−4)), disease-free survival (HR: 5.22, 95% CI: 2.17 – 12.53, P < 0.001 (2.21 * 10−4)), and recurrence-free survival (HR: 1.57, 95% CI: 1.32 – 1.85, P < 0.001 (1.80*10−7)) in various carcinomas. However, results of subgroup analyses revealed that down-regulated miR-9 was associated with poor overall survival (HR: 0.45, 95% CI: 0.28 – 0.73, P < 0.001 (1.13*10−3)) and progress-free survival (HR: 0.46, 95% CI: 0.34 – 0.62, P < 0.001 (5.03*10−7)) in ovarian cancer patients. By subgroup analyses we also found that sample collecting time and patients’ origin had little influence on the result of OS. These results indicate that in most cancer types the highly expressed miR-9 is associated with poor survival of patients, whereas the down-regulated miR-9 may predict poor prognosis in patients with ovarian cancer.
Collapse
Affiliation(s)
- Han Sun
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, P.R. China
| | - Yingjie Shao
- Department of Radiation Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, P.R. China
| | - Jin Huang
- Department of Radiation Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, P.R. China
| | - Siwei Sun
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, P.R. China
| | - Yijie Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, P.R. China
| | - Pinghui Zhou
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, P.R. China
| | - Huilin Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, P.R. China
| |
Collapse
|
9
|
Phase I study of MRX34, a liposomal miR-34a mimic, administered twice weekly in patients with advanced solid tumors. Invest New Drugs 2016; 35:180-188. [PMID: 27917453 DOI: 10.1007/s10637-016-0407-y] [Citation(s) in RCA: 605] [Impact Index Per Article: 75.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 11/09/2016] [Indexed: 12/12/2022]
Abstract
Purpose Naturally occurring tumor suppressor microRNA-34a (miR-34a) downregulates the expression of >30 oncogenes across multiple oncogenic pathways, as well as genes involved in tumor immune evasion, but is lost or under-expressed in many malignancies. This first-in-human, phase I study assessed the maximum tolerated dose (MTD), safety, pharmacokinetics, and clinical activity of MRX34, a liposomal miR-34a mimic, in patients with advanced solid tumors. Patients and Methods Adult patients with solid tumors refractory to standard treatment were enrolled in a standard 3 + 3 dose escalation trial. MRX34 was given intravenously twice weekly (BIW) for three weeks in 4-week cycles. Results Forty-seven patients with various solid tumors, including hepatocellular carcinoma (HCC; n = 14), were enrolled. Median age was 60 years, median prior therapies was 4 (range, 1-12), and most were Caucasian (68%) and male (57%). Most common adverse events (AEs) included fever (all grade %/G3%: 64/2), fatigue (57/13), back pain (57/11), nausea (49/2), diarrhea (40/11), anorexia (36/4), and vomiting (34/4). Laboratory abnormalities included lymphopenia (G3%/G4%: 23/9), neutropenia (13/11), thrombocytopenia (17/0), increased AST (19/4), hyperglycemia (13/2), and hyponatremia (19/2). Dexamethasone premedication was required to manage infusion-related AEs. The MTD for non-HCC patients was 110 mg/m2, with two patients experiencing dose-limiting toxicities of G3 hypoxia and enteritis at 124 mg/m2. The half-life was >24 h, and Cmax and AUC increased with increasing dose. One patient with HCC achieved a prolonged confirmed PR lasting 48 weeks, and four patients experienced SD lasting ≥4 cycles. Conclusion MRX34 treatment with dexamethasone premedication was associated with acceptable safety and showed evidence of antitumor activity in a subset of patients with refractory advanced solid tumors. The MTD for the BIW schedule was 110 mg/m2 for non-HCC and 93 mg/m2 for HCC patients. Additional dose schedules of MRX34 have been explored to improve tolerability.
Collapse
|
10
|
The orphan nuclear receptor NR4A2 is part of a p53-microRNA-34 network. Sci Rep 2016; 6:25108. [PMID: 27121375 PMCID: PMC4848494 DOI: 10.1038/srep25108] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 04/11/2016] [Indexed: 12/13/2022] Open
Abstract
Nuclear receptor subfamily 4 group A member 2 (NR4A2) is an orphan nuclear receptor that is over-expressed in cancer and promotes cell proliferation, migration, transformation, and chemoresistance. Increased expression and function of NR4A2 have been attributed to various signaling pathways, but little is known about microRNA (miRNA) regulation of NR4A2 in cancer. To investigate the posttranscriptional regulation of NR4A2, we used a 3′ untranslated region (UTR) reporter screen and identified miR-34 as a putative regulator of NR4A2. By using computer predictions, we identified and confirmed an miRNA recognition element in the 3′ UTR of NR4A2 that was responsible for miR-34–mediated suppression. We next demonstrated that overexpression of exogenous miR-34 or activation of the p53 pathway, which regulates endogenous miR-34 expression, decreased NR4A2 expression. Consistent with previous reports, overexpression of NR4A2 blocked the induction of p53 target genes, including mir-34a. This was a phenotypic effect, as NR4A2 overexpression could rescue cells from p53-induced inhibition of proliferation. In summary, our results are the first characterization of a cancer-related miRNA capable of regulating NR4A2 and suggest a network and possible feedback mechanism involving p53, miR-34, and NR4A2.
Collapse
|