1
|
Lin H, Luo Y, Gong T, Fang H, Li H, Ye G, Zhang Y, Zhong M. GDF15 induces chemoresistance to oxaliplatin by forming a reciprocal feedback loop with Nrf2 to maintain redox homeostasis in colorectal cancer. Cell Oncol (Dordr) 2024; 47:1149-1165. [PMID: 38386232 DOI: 10.1007/s13402-024-00918-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2024] [Indexed: 02/23/2024] Open
Abstract
PURPOSE Growth differentiating Factor 15 (GDF15) is linked to several cancers, but its effect on chemoresistance in colorectal cancer (CRC) remains unclear. Here, we investigated the role of GDF15 in the chemotherapeutic response of CRC patients to oxaliplatin (L-OHP). METHODS GDF15 levels in serum and tumour tissues were detected in CRC patients have received L-OHP-based neoadjuvant chemotherapy. The effects of GDF15 neutralization or GDF15 knockdown on cell proliferation, apoptosis and intracellular reactive oxygen species (ROS) levels were analysed in vitro and in vivo. Co-immunoprecipitation (Co-IP), Chromatin Immunoprecipitation (ChIP) and luciferase reporter assays were used to explore the interaction between GDF15 and Nrf2. RESULTS In this study, we found that GDF15 alleviates oxidative stress to induce chemoresistance of L-OHP in CRC. Mechanically, GDF15 posttranscriptionally regulates protein stability of Nrf2 through the canonical PI3K/AKT/GSK3β signaling pathway, and in turn, Nrf2 acts as a transcription factor to regulate GDF15 expression to form a positive feedback loop, resulting in the maintenance of redox homeostasis balance in CRC. Furthermore, a positive correlation between GDF15 and Nrf2 was observed in clinical CRC samples, and simultaneous overexpression of both GDF15 and Nrf2 was associated with poor prognosis in CRC patients treated with L-OHP. Simultaneous inhibition of both GDF15 and Nrf2 significantly increases the response to L-OHP in an L-OHP-resistant colorectal cancer cells-derived mouse xenograft model. CONCLUSION This study identified a novel GDF15-Nrf2 positive feedback loop that drives L-OHP resistance and suggested that the GDF15-Nrf2 axis is a potential therapeutic target for the treatment of L-OHP-resistant CRC.
Collapse
Affiliation(s)
- Haiping Lin
- Department of Gastrointestinal Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Luo
- Department of Gastrointestinal Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingyue Gong
- Department of Gastrointestinal Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongsheng Fang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hao Li
- Department of Gastrointestinal Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guangyao Ye
- Department of Gastrointestinal Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Zhang
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiaotong University, Shanghai, China.
| | - Ming Zhong
- Department of Gastrointestinal Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
Mao X, Xu J, Xiao M, Liang C, Hua J, Liu J, Wang W, Yu X, Meng Q, Shi S. ARID3A enhances chemoresistance of pancreatic cancer via inhibiting PTEN-induced ferroptosis. Redox Biol 2024; 73:103200. [PMID: 38781729 PMCID: PMC11145557 DOI: 10.1016/j.redox.2024.103200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 05/03/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024] Open
Abstract
Currently, chemotherapy remains occupying a pivotal place in the treatment of pancreatic ductal adenocarcinoma (PDAC). Nonetheless, the emergence of drug resistance in recent years has limited the clinical efficacy of chemotherapeutic agents, especially gemcitabine (GEM). Through bioinformatics analysis, AT-rich Interactive Domain-containing Protein 3A (ARID3A), one of transcription factors, is discovered to possibly participate in this progress. This study thoroughly investigates the potential role of ARID3A in the malignant progression and GEM chemoresistance of PDAC and explores the underlying mechanisms. The results indicate that ARID3A knockdown suppresses tumor development and enhances the sensitivity of PDAC cells to GEM in vitro and vivo. Mechanically, CUT&Tag profiling sequencing, RNA-sequencing and functional studies demonstrates that decreased ARID3A expression alleviates the transcriptional inhibition of phosphatase and tensin homolog (PTEN), consequently leading to glutathione peroxidase 4 (GPX4) depletion and increased lipid peroxidation levels. Activated ferroptosis induced by the inhibition of GPX4 subsequently restricts tumor progression and reduces GEM resistance in PDAC. This research identifies the ferroptosis regulatory pathway of ARID3A-PTEN-GPX4 axis and reveals its critical role in driving the progression and chemoresistance of pancreatic cancer. Notably, both inhibition of ARID3A and enhancement of ferroptosis can increase chemosensitivity to GEM, which offers a promising opportunity for developing therapeutic strategies to combat acquired chemotherapy resistance in pancreatic cancer.
Collapse
Affiliation(s)
- Xiaoqi Mao
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Mingming Xiao
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Chen Liang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Jie Hua
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Jiang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Qingcai Meng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
3
|
Yue Y, She X, Ding W, Chen S, Xiao Q, Pan B, Zhou L, Yin Y, Li Y, Wang S, Xu M. A novel Senescence-Based prognostic model unveils tumor interactions and drug resistance in colorectal cancer. Int Immunopharmacol 2024; 134:112197. [PMID: 38733826 DOI: 10.1016/j.intimp.2024.112197] [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: 02/17/2024] [Revised: 04/12/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND In China, CRC incidence is escalating. The main hurdles are heterogeneity and drug resistance. This research delves into cellular senescence in CRC, aiming to devise a prognostic model and pinpoint mechanisms impacting drug resistance. METHODS Mendelian randomization (MR) analysis confirmed the association between CRC and cellular aging. The Cancer Genome Atlas (TCGA)-CRC data served as the training set, with GSE38832 and GSE39582 as validation sets. Various bioinformatics methods were employed to construct and validate a risk model. CRC cells with NADPH Oxidase 4 (NOX4) knockout were generated using CRISPR-Cas9 technology. Protein blotting and colony formation assays elucidated the role of NOX4 in CRC cell aging and drug resistance. RESULTS A prognostic model, derived from dataset analysis, uncovered a link between high-risk groups and cancer progression. Notable differences in the tumor microenvironment were observed between risk groups. Finally, NOX4 was found to be linked with aging and drug resistance in CRC. CONCLUSION This research presents a novel senescence-based CRC prognosis model. It identifies NOX4's role in CRC drug resistance, suggesting it is a potential treatment target.
Collapse
Affiliation(s)
- Yanzhe Yue
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, Jiangsu, China; School of Basic-Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, 211198, Nanjing, Jiangsu, China
| | - Xiangjian She
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, Jiangsu, China; School of Basic-Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, 211198, Nanjing, Jiangsu, China
| | - Wenbo Ding
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, Jiangsu, China; School of Basic-Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, 211198, Nanjing, Jiangsu, China
| | - Shuyu Chen
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, Jiangsu, China; School of Basic-Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, 211198, Nanjing, Jiangsu, China
| | - Qianni Xiao
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, Jiangsu, China; School of Basic-Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, 211198, Nanjing, Jiangsu, China
| | - Bei Pan
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, Jiangsu, China
| | - Linpeng Zhou
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, Jiangsu, China; School of Basic-Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, 211198, Nanjing, Jiangsu, China
| | - Yujuan Yin
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, Jiangsu, China
| | - Youyue Li
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, Jiangsu, China
| | - Shukui Wang
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, Jiangsu, China; School of Basic-Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, 211198, Nanjing, Jiangsu, China; Jiangsu Collaborative Innovation Center on Cancer Personalized Medicine, Nanjing Medical University, 211166, Nanjing, Jiangsu, China.
| | - Mu Xu
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, China.
| |
Collapse
|
4
|
Efetov SK, Cao Y, Zou J, Dorogov AY, Paramonova NB, Tsoy LV, Droshneva IV, Fatyanova AS. Metastasis of colorectal cancer to the uterine body and fallopian tube: case report and literature review. J Surg Case Rep 2024; 2024:rjae400. [PMID: 38859968 PMCID: PMC11163451 DOI: 10.1093/jscr/rjae400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 05/29/2024] [Indexed: 06/12/2024] Open
Abstract
Colorectal cancer typically metastasizes to the peritoneum, liver, and lungs. However, metastases to the fallopian tube and uterus are uncommon. This case report delves into this rare occurrence of metastasis and discusses its characteristics, diagnostic methods, and treatments based on an extensive literature review. We present the case of a 61-year-old female patient who underwent her initial hospitalization for da Vinci robotic surgery to address colorectal cancer, stage pT3N0M0. However, during routine postoperative follow-up 6 months later, a localized rectal recurrence was detected. The patient commenced chemoradiotherapy with full response. Subsequently, the patient was readmitted due to pelvic pain again, and a magnetic resonance imaging scan revealed an abnormal mass in the patient's left fallopian tube and uterine corpus, infiltrating the myometrium. Consequently, total hysterectomy with bilateral adnexectomy was performed, along with omentectomy, which confirmed metastatic involvement from rectal cancer upon postoperative pathological examination. This case may inform further diagnosis and treatment of colorectal cancer metastasis to the fallopian tube.
Collapse
Affiliation(s)
- Sergey K Efetov
- Department of Faculty Surgery No.2, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia
| | - Yu Cao
- Department of Faculty Surgery No.2, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia
| | - Jinqi Zou
- Department of Faculty Surgery No.2, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia
| | - Anton Y Dorogov
- Department of Faculty Surgery No.2, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia
| | - Nina B Paramonova
- Insitute for Clinical Morphology and Digital Pathology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia
| | - Larisa V Tsoy
- Insitute for Clinical Morphology and Digital Pathology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia
| | - Inna V Droshneva
- P.A. Herzen Moscow Oncology Research Institute, Branch, Nation Medical Radiology Research Center, Ministry of Health of Russia, Moscow, 119435, Russia
| | - Anastasia S Fatyanova
- Department of Oncology, Radiotherapy and Reconstructive Surgery, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia
| |
Collapse
|
5
|
Perrone G, Rigacci L, Roviello G, Landini I, Fabbri A, Iovino L, Puccini B, Cencini E, Orciuolo E, Bocchia M, Bosi A, Mini E, Nobili S. Validation of single nucleotide polymorphisms potentially related to R-CHOP resistance in diffuse large B-cell lymphoma patients. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:21. [PMID: 38835350 PMCID: PMC11149109 DOI: 10.20517/cdr.2024.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/30/2024] [Accepted: 05/16/2024] [Indexed: 06/06/2024]
Abstract
Aim: Diffuse large B-cell lymphoma (DLBCL) is the most common B-cell non-Hodgkin lymphoma (NHL). Despite the availability of clinical and molecular algorithms applied for the prediction of prognosis, in up to 30%-40% of patients, intrinsic or acquired drug resistance occurs. Constitutional genetics may help to predict R-CHOP resistance. This study aimed to validate previously identified single nucleotide polymorphisms (SNPs) in the literature as potential predictors of R-CHOP resistance in DLBCL patients, SNPs. Methods: Twenty SNPs, involved in R-CHOP pharmacokinetics/pharmacodynamics or other pathobiological processes, were investigated in 185 stage I-IV DLBCL patients included in a multi-institution pharmacogenetic study to validate their previously identified correlations with resistance to R-CHOP. Results: Correlations between rs2010963 (VEGFA gene) and sex (P = 0.046), and rs1625895 (TP53 gene) and stage (P = 0.003) were shown. After multivariate analyses, a concordant effect (i.e., increased risk of disease progression and death) was observed for rs1883112 (NCF4 gene) and rs1800871 (IL10 gene). When patients were grouped according to the revised International Prognostic Index (R-IPI), both these SNPs further discriminated progression-free survival (PFS) and overall survival (OS) of the R-IPI-1-2 subgroup. Overall, patients harboring the rare allele showed shorter PFS and OS compared with wild-type patients. Conclusions: Two out of the 20 study SNPs were validated. Thus, these results support the role of previously identified rs1883112 and rs1800871 in predicting DLBCL resistance to R-CHOP and highlight their ability to further discriminate the prognosis of R-IPI-1-2 patients. These data point to the need to also focus on host genetics for a more comprehensive assessment of DLBCL patient outcomes in future prospective trials.
Collapse
Affiliation(s)
- Gabriele Perrone
- Department of Health Sciences, University of Florence, Florence 50139, Italy
| | - Luigi Rigacci
- Research Unit of Hematology, Department of Medicine and Surgery, Campus Biomedico University, Rome 00128, Italy
| | | | - Ida Landini
- Department of Health Sciences, University of Florence, Florence 50139, Italy
| | - Alberto Fabbri
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, Siena 53100, Italy
| | - Lorenzo Iovino
- Unit of Hematology, Santa Chiara University Hospital, University of Pisa, Pisa 56126, Italy
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109-4433, USA
| | - Benedetta Puccini
- Unit of Hematology, Careggi University-Hospital, Florence 50134, Italy
| | - Emanuele Cencini
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, Siena 53100, Italy
| | - Enrico Orciuolo
- Unit of Hematology, Santa Chiara University Hospital, University of Pisa, Pisa 56126, Italy
| | - Monica Bocchia
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, Siena 53100, Italy
| | - Alberto Bosi
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Enrico Mini
- Department of Health Sciences, University of Florence, Florence 50139, Italy
- Authors contributed equally
| | - Stefania Nobili
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence 50139, Italy
- Authors contributed equally
| |
Collapse
|
6
|
Chen HY, Li XN, Yang L, Ye CX, Chen ZL, Wang ZJ. CircVMP1 promotes glycolysis and disease progression by upregulating HKDC1 in colorectal cancer. ENVIRONMENTAL TOXICOLOGY 2024; 39:1617-1630. [PMID: 38009649 DOI: 10.1002/tox.24061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 10/16/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Circular RNAs (circRNAs) have been reported to play important roles in cancers. Here, we characterized circVMP1 (hsa_circ_0006508), an important circRNA which promoted glycolysis and disease progression in colorectal cancer (CRC). In this study, we aimed to explore the mechanism by which circVMP1 regulated tumor glycolysis and its related pathways in promoting CRC cell proliferation and metastasis. METHODS The expression level of circVMP1 in CRC tissues and adjacent normal tissues was detected using quantitative PCR. In vitro and in vivo functional experiments were used to evaluate the effects of circVMP1 in the regulation of CRC cell proliferation and migration. Mitochondrial stress tests and glycolysis stress tests were conducted to detect the effect of circVMP1 on oxidative phosphorylation and glycolysis. Dual-luciferase reporter and RNA immunoprecipitation assays were used to evaluate the interaction between circVMP1, miR-3167, and HKDC1. RESULTS We demonstrated that the level of circVMP1 was significantly upregulated in CRC tissues compared with normal tissues. In HCT116 and SW480 cells, overexpression of circVMP1 promoted proliferation, metastasis, and glycolysis. In vivo analysis indicated that circVMP1 accelerated the proliferation of xenograft tumors. As for the mechanism, overexpression of circVMP1 increased the levels of hexokinase domain component 1 (HKDC1) through competitive binding with miR-3167. CONCLUSION Our study reported that circVMP1 was one of the tumor driver genes that promoted CRC malignant progression and glycolysis by upregulating HKDC1. CircVMP1/miR-3167/HKDC1 was a signaling axis that might be a target for CRC therapy.
Collapse
Affiliation(s)
- Hong-Yu Chen
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xiang-Nan Li
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Lei Yang
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Chun-Xiang Ye
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zhi-Lei Chen
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zhen-Jun Wang
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Lee SO, Joo SH, Lee JY, Kwak AW, Kim KT, Cho SS, Yoon G, Choi YH, Park JW, Shim JH. Licochalcone C Inhibits the Growth of Human Colorectal Cancer HCT116 Cells Resistant to Oxaliplatin. Biomol Ther (Seoul) 2024; 32:104-114. [PMID: 38148556 PMCID: PMC10762277 DOI: 10.4062/biomolther.2023.167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/12/2023] [Accepted: 10/19/2023] [Indexed: 12/28/2023] Open
Abstract
Licochalcone C (LCC; PubChem CID:9840805), a chalcone compound originating from the root of Glycyrrhiza inflata, has shown anticancer activity against skin cancer, esophageal squamous cell carcinoma, and oral squamous cell carcinoma. However, the therapeutic potential of LCC in treating colorectal cancer (CRC) and its underlying molecular mechanisms remain unclear. Chemotherapy for CRC is challenging because of the development of drug resistance. In this study, we examined the antiproliferative activity of LCC in human colorectal carcinoma HCT116 cells, oxaliplatin (Ox) sensitive and Ox-resistant HCT116 cells (HCT116-OxR). LCC significantly and selectively inhibited the growth of HCT116 and HCT116-OxR cells. An in vitro kinase assay showed that LCC inhibited the kinase activities of EGFR and AKT. Molecular docking simulations using AutoDock Vina indicated that LCC could be in ATP-binding pockets. Decreased phosphorylation of EGFR and AKT was observed in the LCC-treated cells. In addition, LCC induced cell cycle arrest by modulating the expression of cell cycle regulators p21, p27, cyclin B1, and cdc2. LCC treatment induced ROS generation in CRC cells, and the ROS induction was accompanied by the phosphorylation of JNK and p38 kinases. Moreover, LCC dysregulated mitochondrial membrane potential (MMP), and the disruption of MMP resulted in the release of cytochrome c into the cytoplasm and activation of caspases to execute apoptosis. Overall, LCC showed anticancer activity against both Ox-sensitive and Ox-resistant CRC cells by targeting EGFR and AKT, inducing ROS generation and disrupting MMP. Thus, LCC may be potential therapeutic agents for the treatment of Ox-resistant CRC cells.
Collapse
Affiliation(s)
- Seung-On Lee
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
| | - Sang Hoon Joo
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Jin-Young Lee
- Department of Biological Sciences, Keimyung University, Daegu 42601, Republic of Korea
| | - Ah-Won Kwak
- Biosystem Research Group, Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Ki-Taek Kim
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
| | - Seung-Sik Cho
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
| | - Goo Yoon
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Korean Medicine, Dong-Eui University, Busan 47227, Republic of Korea
| | - Jin Woo Park
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
| | - Jung-Hyun Shim
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China
| |
Collapse
|
8
|
Maslarinou A, Manolopoulos VG, Ragia G. Pharmacogenomic-guided dosing of fluoropyrimidines beyond DPYD: time for a polygenic algorithm? Front Pharmacol 2023; 14:1184523. [PMID: 37256234 PMCID: PMC10226670 DOI: 10.3389/fphar.2023.1184523] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 04/19/2023] [Indexed: 06/01/2023] Open
Abstract
Fluoropyrimidines are chemotherapeutic agents widely used for the treatment of various solid tumors. Commonly prescribed FPs include 5-fluorouracil (5-FU) and its oral prodrugs capecitabine (CAP) and tegafur. Bioconversion of 5-FU prodrugs to 5-FU and subsequent metabolic activation of 5-FU are required for the formation of fluorodeoxyuridine triphosphate (FdUTP) and fluorouridine triphosphate, the active nucleotides through which 5-FU exerts its antimetabolite actions. A significant proportion of FP-treated patients develop severe or life-threatening, even fatal, toxicity. It is well known that FP-induced toxicity is governed by genetic factors, with dihydropyrimidine dehydrogenase (DPYD), the rate limiting enzyme in 5-FU catabolism, being currently the cornerstone of FP pharmacogenomics. DPYD-based dosing guidelines exist to guide FP chemotherapy suggesting significant dose reductions in DPYD defective patients. Accumulated evidence shows that additional variations in other genes implicated in FP pharmacokinetics and pharmacodynamics increase risk for FP toxicity, therefore taking into account more gene variations in FP dosing guidelines holds promise to improve FP pharmacotherapy. In this review we describe the current knowledge on pharmacogenomics of FP-related genes, beyond DPYD, focusing on FP toxicity risk and genetic effects on FP dose reductions. We propose that in the future, FP dosing guidelines may be expanded to include a broader ethnicity-based genetic panel as well as gene*gene and gender*gene interactions towards safer FP prescription.
Collapse
Affiliation(s)
- Anthi Maslarinou
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
- Individualised Medicine and Pharmacological Research Solutions Center, Alexandroupolis, Greece
| | - Vangelis G. Manolopoulos
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
- Individualised Medicine and Pharmacological Research Solutions Center, Alexandroupolis, Greece
- Clinical Pharmacology Unit, Academic General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Georgia Ragia
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
- Individualised Medicine and Pharmacological Research Solutions Center, Alexandroupolis, Greece
| |
Collapse
|
9
|
Ren SN, Zhang ZY, Guo RJ, Wang DR, Chen FF, Chen XB, Fang XD. Application of nanotechnology in reversing therapeutic resistance and controlling metastasis of colorectal cancer. World J Gastroenterol 2023; 29:1911-1941. [PMID: 37155531 PMCID: PMC10122790 DOI: 10.3748/wjg.v29.i13.1911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 02/02/2023] [Accepted: 03/21/2023] [Indexed: 04/06/2023] Open
Abstract
Colorectal cancer (CRC) is the most common digestive malignancy across the world. Its first-line treatments applied in the routine clinical setting include surgery, chemotherapy, radiotherapy, targeted therapy, and immunotherapy. However, resistance to therapy has been identified as the major clinical challenge that fails the treatment method, leading to recurrence and distant metastasis. An increasing number of studies have been attempting to explore the underlying mechanisms of the resistance of CRC cells to different therapies, which can be summarized into two aspects: (1) The intrinsic characters and adapted alterations of CRC cells before and during treatment that regulate the drug metabolism, drug transport, drug target, and the activation of signaling pathways; and (2) the suppressive features of the tumor microenvironment (TME). To combat the issue of therapeutic resistance, effective strategies are warranted with a focus on the restoration of CRC cells’ sensitivity to specific treatments as well as reprogramming impressive TME into stimulatory conditions. To date, nanotechnology seems promising with scope for improvement of drug mobility, treatment efficacy, and reduction of systemic toxicity. The instinctive advantages offered by nanomaterials enable the diversity of loading cargoes to increase drug concentration and targeting specificity, as well as offer a platform for trying the combination of different treatments to eventually prevent tumor recurrence, metastasis, and reversion of therapy resistance. The present review intends to summarize the known mechanisms of CRC resistance to chemotherapy, radiotherapy, immunotherapy, and targeted therapy, as well as the process of metastasis. We have also emphasized the recent application of nanomaterials in combating therapeutic resistance and preventing metastasis either by combining with other treatment approaches or alone. In summary, nanomedicine is an emerging technology with potential for CRC treatment; hence, efforts should be devoted to targeting cancer cells for the restoration of therapeutic sensitivity as well as reprogramming the TME. It is believed that the combined strategy will be beneficial to achieve synergistic outcomes contributing to control and management of CRC in the future.
Collapse
Affiliation(s)
- Sheng-Nan Ren
- Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Zhan-Yi Zhang
- Bethune Third Clinical Medical College, Jilin University, Changchun 130021, Jilin Province, China
| | - Rui-Jie Guo
- Bethune Third Clinical Medical College, Jilin University, Changchun 130021, Jilin Province, China
| | - Da-Ren Wang
- Bethune Third Clinical Medical College, Jilin University, Changchun 130021, Jilin Province, China
| | - Fang-Fang Chen
- Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Xue-Bo Chen
- Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Xue-Dong Fang
- Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| |
Collapse
|
10
|
Das A, Adhikari S, Deka D, Baildya N, Sahare P, Banerjee A, Paul S, Bisgin A, Pathak S. An Updated Review on the Role of Nanoformulated Phytochemicals in Colorectal Cancer. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59040685. [PMID: 37109643 PMCID: PMC10143464 DOI: 10.3390/medicina59040685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 04/29/2023]
Abstract
The most common cancer-related cause of death worldwide is colorectal cancer. It is initiated with the formation of polyps, which further cause the development of colorectal cancer in multistep phases. Colorectal cancer mortality is high despite recent treatment breakthroughs and a greater understanding of its pathophysiology. Stress is one of the major causes of triggering different cellular signalling cascades inside the body and which might turn toward the development of cancer. Naturally occurring plant compounds or phytochemicals are being studied for medical purposes. Phytochemicals' benefits are being analyzed for inflammatory illnesses, liver failure, metabolic disorders, neurodegenerative disorders, and nephropathies. Cancer treatment with fewer side effects and better outcomes has been achieved by combining phytochemicals with chemotherapy. Resveratrol, curcumin, and epigallocatechin-3-gallate have been studied for their chemotherapeutic and chemopreventive potentiality, but hydrophobicity, solubility, poor bioavailability, and target selectivity limit the clinical uses of these compounds. The therapeutic potential is maximized by utilizing nanocarriers such as liposomes, micelles, nanoemulsions, and nanoparticles to increase phytochemical bioavailability and target specificity. This updated literature review discusses the clinical limitations, increased sensitivity, chemopreventive and chemotherapeutic effects, and the clinical limitations of the phytochemicals.
Collapse
Affiliation(s)
- Alakesh Das
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| | - Suman Adhikari
- Department of Chemistry, Govt. Degree College, Dharmanagar 799253, India
| | - Dikshita Deka
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| | | | - Padmavati Sahare
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM 3001, Juriquilla 76230, Querétaro, Mexico
| | - Antara Banerjee
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| | - Sujay Paul
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, San Pablo 76130, Querétaro, Mexico
| | - Atil Bisgin
- Cukurova University AGENTEM (Adana Genetic Diseases Diagnosis and Treatment Center), Medical Genetics Department of Medical Faculty, Cukurova University, Adana 01330, Turkey
- InfoGenom RD Laboratories of Cukurova Technopolis, Adana 01330, Turkey
| | - Surajit Pathak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| |
Collapse
|
11
|
Cura Y, Pérez-Ramírez C, Sánchez-Martín A, Membrive-Jimenez C, Valverde-Merino MI, González-Flores E, Morales AJ. Influence of Single-Nucleotide Polymorphisms on Clinical Outcomes of Capecitabine-Based Chemotherapy in Colorectal Cancer Patients: A Systematic Review. Cancers (Basel) 2023; 15:cancers15061821. [PMID: 36980706 PMCID: PMC10046456 DOI: 10.3390/cancers15061821] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
The aim of this systematic review was to provide a comprehensive overview of the literature published in the last decade on the association of single-nucleotide polymorphisms in genes involved in the pharmacodynamic and pharmacokinetic pathways of capecitabine with treatment outcomes among colorectal cancer patients. A systematic search of the literature published in the last 10 years was carried out in two databases (Medline and Scopus) using keywords related to the objective. Quality assessment of the studies included was performed using an assessment tool derived from the Strengthening the Reporting of Genetic Association (STREGA) statement. Thirteen studies were included in this systematic review. Genes involved in bioactivation, metabolism, transport, mechanism of action of capecitabine, DNA repair, and folate cycle were associated with toxicity. Meanwhile, genes related to DNA repair were associated with therapy effectiveness. This systematic review reveals that several SNPs other than the four DPYD variants that are screened in clinical practice could have an impact on treatment outcomes. These findings suggest the identification of future predictive biomarkers of effectiveness and toxicity in colorectal cancer patients treated with capecitabine. However, the evidence is sparse and requires further validation.
Collapse
Affiliation(s)
- Yasmin Cura
- Pharmacy Service, Pharmacogenetics Unit, Hospital Universitario Virgen de las Nieves, Avda. de las Fuerzas Armadas 2, 18004 Granada, Spain
| | - Cristina Pérez-Ramírez
- Department of Biochemistry and Molecular Biology II, José Mataix Institute of Nutrition and Food Technology, Center for Biomedical Research, Universidad de Granada, Avda. del Conocimiento s/n, 18016 Granada, Spain
- Correspondence:
| | - Almudena Sánchez-Martín
- Pharmacy Service, Pharmacogenetics Unit, Hospital Universitario Virgen de las Nieves, Avda. de las Fuerzas Armadas 2, 18004 Granada, Spain
| | - Cristina Membrive-Jimenez
- Pharmacy Service, Pharmacogenetics Unit, Hospital Universitario Virgen de las Nieves, Avda. de las Fuerzas Armadas 2, 18004 Granada, Spain
| | - María Isabel Valverde-Merino
- Pharmaceutical Care Research Group, Facultad de Farmacia, Universidad de Granada, Campus de la Cartuja, 18071 Granada, Spain
| | - Encarnación González-Flores
- Medical Oncology, Hospital Universitario Virgen de las Nieves, Avda. de las Fuerzas Armadas 2, 18004 Granada, Spain
- Biosanitary Research Institute of Granada, Ibs.Granada, Avda. de Madrid, 15, 18012 Granada, Spain
| | - Alberto Jiménez Morales
- Pharmacy Service, Pharmacogenetics Unit, Hospital Universitario Virgen de las Nieves, Avda. de las Fuerzas Armadas 2, 18004 Granada, Spain
| |
Collapse
|
12
|
Hua Q, Lu Y, Wang D, Da J, Peng W, Sun G, Gu K, Wang H, Zhu Y. KIAA1199 promotes oxaliplatin resistance and epithelial mesenchymal transition of colorectal cancer via protein O-GlcNAcylation. Transl Oncol 2023; 28:101617. [PMID: 36610242 PMCID: PMC9850197 DOI: 10.1016/j.tranon.2023.101617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/24/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023] Open
Abstract
Oxaliplatin is a commonly used platinum drug for colorectal cancer (CRC). However, the treatment of CRC by oxaliplatin usually fails because of drug resistance, which results in a huge challenge in the therapy of CRC. Elucidation of molecular mechanisms may help to overcome oxaliplatin resistance of CRC. In our study, we revealed that KIAA1199 can promote oxaliplatin resistance of CRC. Mechanistically, KIAA1199 prevents oxaliplatin mediated apoptosis via up-regulated PARP1 derived from reduced endoplasmic reticulum stress induced by protein O-GlcNAcylation. In the meantime, KIAA1199 can also trigger epithelial mesenchymal transition by stabilizing SNAI1 protein via O-GlcNAcylation. Therefore, KIAA1199 has great potential to be a novel biomarker, therapeutic target for oxaliplatin resistance and metastasis of CRC.
Collapse
Affiliation(s)
- Qingling Hua
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China
| | - Yuanyuan Lu
- Department of Radiation Oncology, The First Affiliated Hospital of Wannan Medical College, Wuhu 241004, China
| | - Dingxiang Wang
- Department of Psychology, The fourth people's hospital, Wuhu, 241003, China
| | - Jie Da
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China
| | - Wanren Peng
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China
| | - Guoping Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China
| | - Kangsheng Gu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China
| | - Yanzhe Zhu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China,Corresponding author.
| |
Collapse
|
13
|
Ghatak S, Hascall VC, Karamanos N, Markwald RR, Misra S. Chemotherapy induces feedback up-regulation of CD44v6 in colorectal cancer initiating cells through β-catenin/MDR1 signaling to sustain chemoresistance. Front Oncol 2022; 12:906260. [PMID: 36330477 PMCID: PMC9623568 DOI: 10.3389/fonc.2022.906260] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/15/2022] [Indexed: 08/05/2023] Open
Abstract
Chemoresistance in colorectal cancer initiating cells (CICs) involves the sustained activation of multiple drug resistance (MDR) and WNT/β-catenin signaling pathways, as well as of alternatively spliced-isoforms of CD44 containing variable exon-6 (CD44v6). In spite of its importance, mechanisms underlying the sustained activity of WNT/β-catenin signaling have remained elusive. The presence of binding elements of the β-catenin-interacting transcription factor TCF4 in the MDR1 and CD44 promoters suggests that crosstalk between WNT/β-catenin/TCF4-activation and the expression of the CD44v6 isoform mediated by FOLFOX, a first-line chemotherapeutic agent for colorectal cancer, could be a fundamental mechanism of FOLFOX resistance. Our results identify that FOLFOX treatment induced WNT3A secretion, which stimulated a positive feedback loop coupling β-catenin signaling and CD44v6 splicing. In conjunction with FOLFOX induced WNT3A signal, specific CD44v6 variants produced by alternative splicing subsequently enhance the late wave of WNT/β-catenin activation to facilitate cell cycle progression. Moreover, we revealed that FOLFOX-mediated sustained WNT signal requires the formation of a CD44v6-LRP6-signalosome in caveolin microdomains, which leads to increased FOLFOX efflux. FOLFOX-resistance in colorectal CICs occurs in the absence of tumor-suppressor disabled-2 (DAB2), an inhibitor of WNT/β-catenin signaling. Conversely, in sensitive cells, DAB2 inhibition of WNT-signaling requires interaction with a clathrin containing CD44v6-LRP6-signalosome. Furthermore, full-length CD44v6, once internalized through the caveolin-signalosome, is translocated to the nucleus where in complex with TCF4, it binds to β-catenin/TCF4-regulated MDR1, or to CD44 promoters, which leads to FOLFOX-resistance and CD44v6 transcription through transcriptional-reprogramming. These findings provide evidence that targeting CD44v6-mediated LRP6/β-catenin-signaling and drug efflux may represent a novel approach to overcome FOLFOX resistance and inhibit tumor progression in colorectal CICs. Thus, sustained drug resistance in colorectal CICs is mediated by overexpression of CD44v6, which is both a functional biomarker and a therapeutic target in colorectal cancer.
Collapse
Affiliation(s)
- Shibnath Ghatak
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
- Department Natural Sciences, Trident Technical College, North Charleston, SC, United States
| | - Vincent C. Hascall
- Department of Biomedical Engineering/ND20, Cleveland Clinic, Cleveland, OH, United States
| | - Nikos Karamanos
- University of Patras, Matrix Pathobiology Res. Group, Department of Chemistry, Patras, Greece
| | - Roger R. Markwald
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Suniti Misra
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
- Department Natural Sciences, Trident Technical College, North Charleston, SC, United States
| |
Collapse
|
14
|
Huang CZ, Zhou Y, Tong QS, Duan QJ, Zhang Q, Du JZ, Yao XQ. Precision medicine-guided co-delivery of ASPN siRNA and oxaliplatin by nanoparticles to overcome chemoresistance of colorectal cancer. Biomaterials 2022; 290:121827. [DOI: 10.1016/j.biomaterials.2022.121827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 09/18/2022] [Accepted: 09/24/2022] [Indexed: 11/02/2022]
|
15
|
Bignucolo A, Scarabel L, Toffoli G, Cecchin E, De Mattia E. Predicting drug response and toxicity in metastatic colorectal cancer: the role of germline markers. Expert Rev Clin Pharmacol 2022; 15:689-713. [PMID: 35829762 DOI: 10.1080/17512433.2022.2101447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Despite the introduction of targeted agents leading to therapeutic advances, clinical management of patients with metastatic colorectal cancer (mCRC) is still challenged by significant interindividual variability in treatment outcomes, both in terms of toxicity and therapy efficacy. The study of germline genetic variants could help to personalize and optimize therapeutic approaches in mCRC. AREAS COVERED A systematic review of pharmacogenetic studies in mCRC patients published on PubMed between 2011 and 2021, evaluating the role of germline variants as predictive markers of toxicity and efficacy of drugs currently approved for treatment of mCRC, was perfomed. EXPERT OPINION Despite the large amount of pharmacogenetic data published to date, only a few genetic markers (i.e., DPYD and UGT1A1 variants) reached the clinical practice, mainly to prevent the toxic effects of chemotherapy. The large heterogeneity of available studies represents the major limitation in comparing results and identifying potential markers for clinical use, the role of which remains exploratory in most cases. However, the available published findings are an important starting point for future investigations. They highlighted new promising pharmacogenetic markers within the network of inflammatory and immune response signaling. In addition, the emerging role of previously overlooked rare variants has been pointed out.
Collapse
Affiliation(s)
- Alessia Bignucolo
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via Franco Gallini 2, 33081 Aviano (PN), Italy
| | - Lucia Scarabel
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via Franco Gallini 2, 33081 Aviano (PN), Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via Franco Gallini 2, 33081 Aviano (PN), Italy
| | - Erika Cecchin
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via Franco Gallini 2, 33081 Aviano (PN), Italy
| | - Elena De Mattia
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Via Franco Gallini 2, 33081 Aviano (PN), Italy
| |
Collapse
|
16
|
Liu X, Zhang Y, Wu X, Xu F, Ma H, Wu M, Xia Y. Targeting Ferroptosis Pathway to Combat Therapy Resistance and Metastasis of Cancer. Front Pharmacol 2022; 13:909821. [PMID: 35847022 PMCID: PMC9280276 DOI: 10.3389/fphar.2022.909821] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/20/2022] [Indexed: 01/18/2023] Open
Abstract
Ferroptosis is an iron-dependent regulated form of cell death caused by excessive lipid peroxidation. This form of cell death differed from known forms of cell death in morphological and biochemical features such as apoptosis, necrosis, and autophagy. Cancer cells require higher levels of iron to survive, which makes them highly susceptible to ferroptosis. Therefore, it was found to be closely related to the progression, treatment response, and metastasis of various cancer types. Numerous studies have found that the ferroptosis pathway is closely related to drug resistance and metastasis of cancer. Some cancer cells reduce their susceptibility to ferroptosis by downregulating the ferroptosis pathway, resulting in resistance to anticancer therapy. Induction of ferroptosis restores the sensitivity of drug-resistant cancer cells to standard treatments. Cancer cells that are resistant to conventional therapies or have a high propensity to metastasize might be particularly susceptible to ferroptosis. Some biological processes and cellular components, such as epithelial–mesenchymal transition (EMT) and noncoding RNAs, can influence cancer metastasis by regulating ferroptosis. Therefore, targeting ferroptosis may help suppress cancer metastasis. Those progresses revealed the importance of ferroptosis in cancer, In order to provide the detailed molecular mechanisms of ferroptosis in regulating therapy resistance and metastasis and strategies to overcome these barriers are not fully understood, we described the key molecular mechanisms of ferroptosis and its interaction with signaling pathways related to therapy resistance and metastasis. Furthermore, we summarized strategies for reversing resistance to targeted therapy, chemotherapy, radiotherapy, and immunotherapy and inhibiting cancer metastasis by modulating ferroptosis. Understanding the comprehensive regulatory mechanisms and signaling pathways of ferroptosis in cancer can provide new insights to enhance the efficacy of anticancer drugs, overcome drug resistance, and inhibit cancer metastasis.
Collapse
Affiliation(s)
- Xuan Liu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yiqian Zhang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Xuyi Wu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu, China
| | - Fuyan Xu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Hongbo Ma
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Mengling Wu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Xia
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu, China
- *Correspondence: Yong Xia,
| |
Collapse
|
17
|
Liu G, Xiao X, Xia Y, Huang W, Chen W, Xu J, Chen S, Wang H, Wei J, Li H, Shu M, Lu X, Zhang C, He Y. Organoids From Mucinous Appendiceal Adenocarcinomas as High-Fidelity Models for Individual Therapy. Front Med (Lausanne) 2022; 9:829033. [PMID: 35721089 PMCID: PMC9201037 DOI: 10.3389/fmed.2022.829033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 04/25/2022] [Indexed: 12/29/2022] Open
Abstract
Background Mucinous appendiceal adenocarcinoma (MAA) is a rare, heterogeneous disease. Patients with unrespectable mucinous appendiceal adenocarcinoma presenting with peritoneal spread are treated by intraperitoneal chemotherapy, hyperthermic intraperitoneal chemotherapy, systemic chemotherapy, or targeted therapy. However, there are no guidelines for efficacious drugs against mucinous appendiceal adenocarcinoma. Therefore, relevant high-fidelity models should be investigated to identify effective drugs for individual therapy. Methods Surgical tumor specimens were obtained from a mucinous appendiceal adenocarcinoma patient. The tissue was digested and organoid culture was established. H&E and immunohistochemistry staining as well as DNA sequencing was performed on tissue and organoid. The pathological characteristics and gene mutations of the organoid were compared to those of the original tumor. Drug sensitivity tests were performed on organoid and the patient clinical responds to chemotherapy and targeted therapy was compared. Results Organoids were successfully established and stably passaged. Pathological characteristics of organoids including H&E staining and expression of protein markers (CK20, CDX-2, STAB2, CD7, PAX8) were consistent to those of the original tumor. Moreover, the organoids carried the same gene mutations as the primary tumor. Sensitivity of the organoids to chemotherapeutic drugs and tyrosine kinase inhibitors included: 5-FU (IC50 43.95 μM), Oxaliplatin (IC50 23.49 μM), SN38 (IC50 1.02 μM), Apatinib (IC50 0.10 μM), Dasatinib (IC50 2.27 μM), Docetaxel (IC50 5.26 μM), Regorafenib (IC50 18.90 μM), and Everolimus (IC50 9.20 μM). The sensitivities of organoid to these drugs were comparable to those of the patient's clinical responses. Conclusion The mucinous appendiceal adenocarcinoma organoid model which retained the characteristics of the primary tumor was successfully established. Combined organoid-based drug screening and high throughput sequencing provided a promising way for mucinous appendiceal adenocarcinoma treatment.
Collapse
Affiliation(s)
- Guangyao Liu
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xing Xiao
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yujian Xia
- Department of Gastrointestinal Surgery, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Weibing Huang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Wei Chen
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Jiannan Xu
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Songyao Chen
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Huijin Wang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Jitao Wei
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Huan Li
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Man Shu
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaofang Lu
- Department of Pathology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Changhua Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yulong He
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.,Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
18
|
Zamami Y, Niimura T, Kawashiri T, Goda M, Naito Y, Fukushima K, Ushio S, Aizawa F, Hamano H, Okada N, Yagi K, Miyata K, Takechi K, Chuma M, Koyama T, Kobayashi D, Shimazoe T, Fujino H, Izawa-Ishizawa Y, Ishizawa K. Identification of prophylactic drugs for oxaliplatin-induced peripheral neuropathy using big data. Biomed Pharmacother 2022; 148:112744. [PMID: 35240525 DOI: 10.1016/j.biopha.2022.112744] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 02/06/2022] [Accepted: 02/18/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Drug repositioning is a cost-effective method to identify novel disease indications for approved drugs; it requires a shorter developmental period than conventional drug discovery methods. We aimed to identify prophylactic drugs for oxaliplatin-induced peripheral neuropathy by drug repositioning using data from large-scale medical information and life science information databases. METHODS Herein, we analyzed the reported data between 2007 and 2017 retrieved from the FDA's database of spontaneous adverse event reports (FAERS) and the LINCS database provided by the National Institute of Health. The efficacy of the drug candidates for oxaliplatin-induced peripheral neuropathy obtained from the database analysis was examined using a rat model of peripheral neuropathy. Additionally, we compared the incidence of peripheral neuropathy in patients who received oxaliplatin at the Tokushima University Hospital, Japan. The effects of statins on the animal model were examined in six-week-old male Sprague-Dawley rats and seven or eight-week-old male BALB/C mice. Retrospective medical chart review included clinical data from Tokushima University Hospital from April 2009 to March 2018. RESULTS Simvastatin, indicated for dyslipidemia, significantly reduced the severity of peripheral neuropathy and oxaliplatin-induced hyperalgesia. In the nerve tissue of model rats, the mRNA expression of Gstm1 increased with statin administration. A retrospective medical chart review using clinical data revealed that the incidence of peripheral neuropathy decreased with statin use. CONCLUSION AND RELEVANCE Thus, drug repositioning using data from large-scale basic and clinical databases enables the discovery of new indications for approved drugs with a high probability of success.
Collapse
Affiliation(s)
- Yoshito Zamami
- Department of Clinical Pharmacy, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan; Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Takahiro Niimura
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Takehiro Kawashiri
- Department of Clinical Pharmacy and Pharmaceutical Care, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Mitsuhiro Goda
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yutaro Naito
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Keijo Fukushima
- Department of Pharmacology for Life Sciences, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Soichiro Ushio
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Fuka Aizawa
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Hirofumi Hamano
- Clinical Research Center for Developmental Therapeutics, Tokushima University Hospital, Tokushima, Japan
| | - Naoto Okada
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Kenta Yagi
- Clinical Research Center for Developmental Therapeutics, Tokushima University Hospital, Tokushima, Japan
| | - Koji Miyata
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kenshi Takechi
- Department of Drug Information Analysis, College of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Japan
| | - Masayuki Chuma
- Department of Hospital Pharmacy and Pharmacology, Asahikawa Medical University, Asahikawa, Japan
| | - Toshihiro Koyama
- Department of Pharmaceutical Biomedicine, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Daisuke Kobayashi
- Department of Clinical Pharmacy and Pharmaceutical Care, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Takao Shimazoe
- Department of Clinical Pharmacy and Pharmaceutical Care, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiromichi Fujino
- Department of Pharmacology for Life Sciences, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yuki Izawa-Ishizawa
- Department of Pharmacology, University of Tokushima Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Keisuke Ishizawa
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan; Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan.
| |
Collapse
|
19
|
Tsai HL, Chen YC, Yin TC, Su WC, Chen PJ, Chang TK, Li CC, Huang CW, Wang JY. Comparison of UGT1A1 polymorphism as guidance of irinotecan dose escalation in RAS wild type metastatic colorectal cancer patients treated with cetuximab or bevacizumab plus FOLFIRI as the first-line therapy. Oncol Res 2022; 29:47-61. [PMID: 35177165 PMCID: PMC9110692 DOI: 10.3727/096504022x16451187313084] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) polymorphism plays a crucial role in the increased susceptibility and toxicity of patients to irinotecan. This retrospective, observational study compared the clinical outcomes and adverse events (AEs) in RAS wild-type metastatic colorectal cancer (mCRC) patients treated with cetuximab or bevacizumab plus FOLFIRI with UGT1A1 genotyping and irinotecan dose escalation as the first-line therapy. In total, 173 patients with mCRC with RAS wild-type were enrolled. Among them, 98 patients were treated with cetuximab, whereas 75 patients were treated with bevacizumab. All patients received irinotecan dose escalation based on UGT1A1 genotyping. We compared the progression-free survival (PFS), overall survival (OS), objective response rates (ORRs), disease control rates (DCRs), metastatectomy, and severe adverse events (SAEs) between the two groups. The clinical effects of primary tumor sidedness and target therapy crossover were further analyzed. Over a median follow-up of 23.0 months [interquartile range (IQR), 15.0–32.5 months], no significant differences were observed between the cetuximab and bevacizumab groups in PFS [18.0 months vs. 14.0 months; 95% confidence interval (CI), 0.517–1.027; hazard ratio (HR), 0.729; p = 0.071], OS (40.0 months vs. 30.0 months; 95% CI, 0.410–1.008; HR, 0.643; p = 0.054), ORR (65.3% vs. 62.7%; p = 0.720), DCR (92.8% vs. 86.7%; p = 0.175), metastatectomy (36.7% vs. 29.3%; p = 0.307), and SAEs (p = 0.685). Regardless of primary tumor sidedness and target therapy crossover, no significant differences were noted in efficacy and safety between the two groups (all p > 0.05). Our results revealed that patients with wild-type RAS mCRC, regardless of biologics, with UGT1A1 genotyping can tolerate escalated doses of irinotecan and potentially achieve a more favorable clinical outcome without significantly increased toxicity.
Collapse
Affiliation(s)
- Hsiang-Lin Tsai
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yen-Cheng Chen
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | - Wei-Chih Su
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Po-Jung Chen
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tsung-Kun Chang
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Chun Li
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Wen Huang
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jaw-Yuan Wang
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
20
|
Qin S, Xu RH, Shen L, Xu J, Bai Y, Yang L, Deng Y, Chen ZD, Zhong H, Pan H, Guo W, Shu Y, Yuan Y, Zhou J, Xu N, Liu T, Ma D, Wu C, Cheng Y, Chen D, Li W, Sun S, Yu Z, Cao P, Chen H, Wang J, Wang S, Wang H, Wang N, Zhang B, Zhang Q, Su W, Guo X, Li J. Subgroup Analysis by Liver Metastasis in the FRESCO Trial Comparing Fruquintinib versus Placebo Plus Best Supportive Care in Chinese Patients with Metastatic Colorectal Cancer. Onco Targets Ther 2021; 14:4439-4450. [PMID: 34408440 PMCID: PMC8364970 DOI: 10.2147/ott.s307273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022] Open
Abstract
Objective The aim of the present subgroup analysis of the FRESCO trial is to determine the efficacy and hepatotoxicity of fruquintinib in Chinese patients with metastatic CRC with liver metastasis (CRLM) who were receiving third-line or posterior-line therapy. Methods Overall survival (OS) and progression-free survival (PFS) were evaluated by Kaplan–Meier method. Hazard ratio (HR) was estimated through Cox proportional hazards model. Hepatotoxicity was coded using the standardized MedDRA queries of hepatic failure, fibrosis, cirrhosis, and other liver injury-related conditions and graded using the Common Terminology Criteria Adverse Events grades. The efficacy of fruquintinib in patients with CRLM was evaluated in various subgroups. Results A total of 287 (69.0%) patients with metastatic CRC had liver metastasis (LM, fruquintinib: 185 and placebo: 102). Median OS in patients with CRLM was significantly prolonged with fruquintinib compared with placebo (8.61 months vs 5.98 months; HR=0.59, 95% CI, 0.45–0.77, P<0.001). In patients with CRLM, the incremental median PFS for patients in the fruquintinib-treated group was significantly higher than in the placebo group (median PFS: 3.71 vs.1.84 months; HR=0.22, 95% CI: 0.17–0.30; P<0.001). Compared with placebo, significant improvements in OS were observed with fruquintinib in LM patients regardless of lung metastasis, prior target therapy, and K-RAS status. In patients with CRLM, treatment-emergent hepatotoxicities of any grade occurred in 7 (3.8%) patients in the fruquintinib group vs 2 (2.0%) in the placebo group. Conclusion Fruquintinib demonstrated a statistically significant increase in OS and PFS as compared with placebo in Chinese patients with CRLM. The hepatotoxicity of fruquintinib was less reported, and comparable with placebo in patients with CRLM. ClinicalTrials.gov Identifier NCT02314819.
Collapse
Affiliation(s)
- Shukui Qin
- Cancer Center, Jinling Hospital, Nanjing, People's Republic of China
| | - Rui-Hua Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Jianming Xu
- Department of Medical Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Yuxian Bai
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - Lei Yang
- Department of Medical Oncology, Nantong Cancer Hospital, Nantong, People's Republic of China
| | - Yanhong Deng
- Department of Medical Oncology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhen-Dong Chen
- Department of Medical Oncology, The Second Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Haijun Zhong
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Weijian Guo
- Department of Medical Oncology, Shanghai Medical College, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China
| | - Yongqian Shu
- Department of Medical Oncology, Jiangsu Provincial Hospital, Nanjing, People's Republic of China
| | - Ying Yuan
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jianfeng Zhou
- Department of Medical Oncology, Peking Union Medical College Hospital, Beijing, People's Republic of China
| | - Nong Xu
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, People's Republic of China
| | - Tianshu Liu
- Department of Medical Oncology, Fudan University Zhongshan Hospital, Shanghai Medical College, Shanghai, People's Republic of China
| | - Dong Ma
- Department of Medical Oncology, Guangdong Provincial People's Hospital, Guangzhou, People's Republic of China
| | - Changping Wu
- Department of Medical Oncology, The First People's Hospital of Changzhou, Changzhou, People's Republic of China
| | - Ying Cheng
- Department of Medical Oncology, Jilin Province Cancer Hospital, Changchun, People's Republic of China
| | - Donghui Chen
- Department of Medical Oncology, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, People's Republic of China
| | - Wei Li
- Department of Medical Oncology, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Sanyuan Sun
- Department of Medical Oncology, Xuzhou Central Hospital, Xuzhou, People's Republic of China
| | - Zhuang Yu
- Department of Medical Oncology, The Affiliated Hospital of Medical College Qingdao University, Qingdao, People's Republic of China
| | - Peiguo Cao
- Department of Medical Oncology, The Third Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Haihui Chen
- Department of Medical Oncology, Liuzhou Worker's Hospital, Liuzhou, People's Republic of China
| | - Jiejun Wang
- Department of Medical Oncology, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai, People's Republic of China
| | - Shubin Wang
- Department of Medical Oncology, Peking University Shenzhen Hospital, Beijing University, Shenzhen, People's Republic of China
| | - Hongbing Wang
- Department of Medical Oncology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou Medical College, Xuzhou, People's Republic of China
| | - Ning Wang
- Eli Lilly and Company, Shanghai, People's Republic of China
| | - Bin Zhang
- Eli Lilly and Company, Shanghai, People's Republic of China
| | - Qiang Zhang
- Eli Lilly and Company, Shanghai, People's Republic of China
| | - Weiguo Su
- Hutchison MediPharma Limited, Shanghai, People's Republic of China
| | - Xiaojun Guo
- Hutchison MediPharma Limited, Shanghai, People's Republic of China
| | - Jin Li
- Department of Medical Oncology, Tongji University Shanghai East Hospital, Shanghai, People's Republic of China
| |
Collapse
|
21
|
Wang G, Wang Y, Yang X, Zhang Y, Lu Y, Li Y. The expression and diagnostic value of serum levels of EphA2 and VEGF-A in patients with colorectal cancer. Cancer Biomark 2021; 31:399-408. [PMID: 34092605 DOI: 10.3233/cbm-201745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Several molecules are highly expressed in the serum of cancer patients, and can be used as serological markers. This approach has become one of the important auxiliary diagnostic methods for cancer. AIM To investigate the correlation between the serum levels of EphA2 and VEGF-A and the pathogenesis of colorectal cancer (CRC) as well as the potential value of these molecules in the diagnosis of CRC. METHODS ELISA was used to detect the levels of EphA2 and VEGF-A in the peripheral venous serum of 106 newly diagnosed patients with CRC and 69 normal controls. The relationship between the serum EphA2 and VEGF-A levels and the clinicopathological characteristics of CRC patients was analyzed. ROC analysis was used to investigate the diagnostic value of the serum EphA2 and VEGF-A levels in CRC, and the optimal cutoff value was calculated. RESULTS The serum levels of EphA2 and VEGF-A in the CRC group were higher than those in the control as well as CEA, the serum level of EphA2 was positively correlated with the VEGF-A levels, but neither was significantly associated with the clinicopathological parameters of CRC. The ROC curve showed that the single index AUC was < 0.7 except for VEGF-A, and the accuracy of the combined diagnosis was higher than that of any other single index. The diagnosis scheme involving all three markers was the best (the sensitivity was 60.40%, the specificity was 92.8%, and the accuracy was 53.1%). The best critical values calculated were EphA2 > 297.92 ng/ml, EphA2 > 183.92 pg/ml and CEA > 5.19 ng/ml. CONCLUSION The serum levels of EphA2 and VEGF-A are high in CRC patients, and the combine detection of CEA, EphA2 and VEGF-A can significantly improve the diagnostic accuracy of CRC.
Collapse
Affiliation(s)
- Ganbiao Wang
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,Department of General Surgery, Guzhen County Traditional Chinese Medicine Hospital, Guzhen, Bengbu, Anhui, China.,Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yigao Wang
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xiaodong Yang
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yaqin Zhang
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yida Lu
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yongxiang Li
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
22
|
Ren Y, Deng R, Cai R, Lu X, Luo Y, Wang Z, Zhu Y, Yin M, Ding Y, Lin J. TUSC3 induces drug resistance and cellular stemness via Hedgehog signaling pathway in colorectal cancer. Carcinogenesis 2021; 41:1755-1766. [PMID: 32338281 DOI: 10.1093/carcin/bgaa038] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 04/08/2020] [Accepted: 04/16/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor suppressor candidate 3 (TUSC3) is a coding gene responsible for N-glycosylation of many critical proteins. TUSC3 gene plays an oncogenic role in colorectal cancer (CRC), however, the role of TUSC3 in drug resistance of CRC is still unclear. The aim of this study is to investigate the biological function and molecular mechanism of TUSC3 in CRC drug resistance. The expression of TUSC3 in CRC is positively correlated to tumor stage in 90 paired clinical samples, and negatively associated with overall survival and disease-free survival of CRC patients. In vitro, TUSC3 promotes the formation of stemness and induces the drug resistance to 5-fluorouracil and cis-dichlorodiammineplatinum(II) in CRC cells. The tissue microarray assay and bioinformatic analysis indicate that TUSC3 may promote the expression of CD133 and ABCC1 via Hedgehog signaling pathway. Treatment of Hedgehog signaling pathway agonist or inhibitor in TUSC3-silenced or TUSC3-overexpressed cells reverse the effects of TUSC3 in cellular stemness phenotype and drug resistance. Meanwhile, coimmunoprecipitation and immunofluorescence assays indicate a tight relationship between TUSC3 and SMO protein. Our data suggest that TUSC3 promotes the formation of cellular stemness and induces drug resistance via Hedgehog signaling pathway in CRC.
Collapse
Affiliation(s)
- Yansong Ren
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou, Guangdong Province, PR China
| | - Ruxia Deng
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou, Guangdong Province, PR China
| | - Rui Cai
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou, Guangdong Province, PR China
| | - Xiansheng Lu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou, Guangdong Province, PR China
| | - Yuejun Luo
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou, Guangdong Province, PR China
| | - Ziyuan Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou, Guangdong Province, PR China
| | - Yuchen Zhu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou, Guangdong Province, PR China
| | - Mengyuan Yin
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou, Guangdong Province, PR China
| | - Yanqing Ding
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou, Guangdong Province, PR China
| | - Jie Lin
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, PR China.,Department of Pathology, Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou, Guangdong Province, PR China
| |
Collapse
|
23
|
IL15RA and SMAD3 Genetic Variants Predict Overall Survival in Metastatic Colorectal Cancer Patients Treated with FOLFIRI Therapy: A New Paradigm. Cancers (Basel) 2021; 13:cancers13071705. [PMID: 33916844 PMCID: PMC8038482 DOI: 10.3390/cancers13071705] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/25/2021] [Accepted: 04/01/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary There is an increasing scientific interest in the study of the interaction between the immune system and drugs in cancer that can affect the efficacy of an anti-cancer treatment. This study was undertaken to better understand if the genetic characteristic of a cancer patient’s immune system can predict the tumor response to the treatment and the duration of survival. The topic was studied on 335 metastatic colorectal cancer patients treated with a first-line chemotherapy (FOLFIRI regimen, irinotecan-5-fluorouracil-leucovorin). The research highlighted two markers, IL15RA-rs7910212 and SMAD3-rs7179840, significantly associated with the patient’s survival. When considering IL15RA-rs7910212 and SMAD3-rs7179840 in combination with other two genetic markers previously investigated (NR1I2-rs1054190, VDR-rs7299460), we built up a highly predictive genetic score of survival. The herein identified markers must be further validated, but still represent good candidates to understand how much a patient with a metastatic colorectal cancer can benefit from a chemotherapy with FOLFIRI regimen. Abstract A new paradigm in cancer chemotherapy derives from the interaction between chemotherapeutics, including irinotecan and 5-fluorouracil (5-FU), and the immune system. The patient’s immune response can modulate chemotherapy effectiveness, and, on the other hand, chemotherapeutic agents can foster tumor cell immunogenicity. On these grounds, the analysis of the cancer patients’ immunogenetic characteristics and their effect on survival after chemotherapy represent a new frontier. This study aims to identify genetic determinants in the immuno-related pathways predictive of overall survival (OS) after FOLFIRI (irinotecan, 5-FU, leucovorin) therapy. Two independent cohorts comprising a total of 335 patients with metastatic colorectal cancer (mCRC) homogeneously treated with first-line FOLFIRI were included in the study. The prognostic effect of 192 tagging genetic polymorphisms in 34 immune-related genes was evaluated using the bead array technology. The IL15RA rs7910212-C allele was associated with worse OS in both discovery (HR: 1.57, p = 0.0327, Bootstrap p-value = 0.0280) and replication (HR: 1.71, p = 0.0411) cohorts. Conversely, SMAD3 rs7179840-C allele was associated with better OS in both discovery (HR: 0.65, p = 0.0202, Bootstrap p-value = 0.0203) and replication (HR: 0.61, p = 0.0216) cohorts. A genetic prognostic score was generated integrating IL15RA-rs7910212 and SMAD3-rs7179840 markers with inflammation-related prognostic polymorphisms we previously identified in the same study population (i.e., PXR [NR1I2]-rs1054190, VDR-rs7299460). The calculated genetic score successfully discriminated patients with different survival probabilities (p < 0.0001 log-rank test). These findings provide new insight on the prognostic value of genetic determinants, such as IL15RA and SMAD3 markers, and could offer a new decision tool to improve the clinical management of patients with mCRC receiving FOLFIRI.
Collapse
|
24
|
Yang C, Zhang Y, Lin S, Liu Y, Li W. Suppressing the KIF20A/NUAK1/Nrf2/GPX4 signaling pathway induces ferroptosis and enhances the sensitivity of colorectal cancer to oxaliplatin. Aging (Albany NY) 2021; 13:13515-13534. [PMID: 33819186 PMCID: PMC8202845 DOI: 10.18632/aging.202774] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 12/18/2020] [Indexed: 04/11/2023]
Abstract
Oxaliplatin resistance can develop in colorectal cancer (CRC), which may involve inhibition of ferroptosis, although further research is needed to understand this potential mechanism. We evaluated CRC cells with acquired oxaliplatin resistance (HCT116-Or) or congenital resistance (H716) to determine whether a ferroptosis inducer (RSL3) or inhibitor (liproxstatin-1) could modulate the effects of oxaliplatin. The results suggested that induction of ferroptosis could significantly reverse the oxaliplatin resistance of the CRC cells. Bioinformatic and cytobiological searches also revealed that KIF20A was highly expressed in the oxaliplatin-resistant cell lines and was strongly correlated with survival among CRC patients. Silencing KIF20A enhanced cellular sensitivity to oxaliplatin both in vivo and in vitro, and silencing KIF20A also suppressed NUAK1 activation, while a NUAK1 agonist (ETC-1002) could reverse the oxaliplatin sensitivity of KIF20A-silenced cells. Moreover, silencing NUAK1 up-regulated the expression of PP1β, down-regulated the phosphorylation of downstream GSK3βSer9, suppressed the nuclear import of Nrf2, inhibited the expression of a ferroptosis key negative regulatory protein (GPX4), and blocked cellular resistance. Applying a Nrf2 agonist (oltipraz) also reversed the oxaliplatin sensitivity of NUAK1-silenced cells. Therefore, cellular ferroptosis may be inhibited via the KIF20A/NUAK1/PP1β/GPX4 pathway in CRC cells, which may underly the resistance of CRC to oxaliplatin.
Collapse
Affiliation(s)
- Changshun Yang
- Department of Surgical Oncology, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Yu Zhang
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China
| | - Shengtao Lin
- Department of Surgical Oncology, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Yi Liu
- Department of Endoscopy, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100000, China
| | - Weihua Li
- Department of Surgical Oncology, Fujian Provincial Hospital, Fuzhou 350001, China
| |
Collapse
|
25
|
MEHDIZADEHTAPEH L, OBAKAN YERLİKAYA P. Endoplasmic reticulum stress and oncomir-associated chemotherapeutic drug resistance mechanisms in breast cancer tumors. Turk J Biol 2021; 45:1-16. [PMID: 33597817 PMCID: PMC7877716 DOI: 10.3906/biy-2010-62] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 01/04/2021] [Indexed: 01/11/2023] Open
Abstract
Breast cancer, as a heterogenous malign disease among the top five leading causes of cancer death worldwide, is defined as by far the most common malignancy in women. It contributes to 25% of all cancer-associated deaths after menopause. Breast cancer is categorized based on the expression levels of cell surface and intracellular steroid receptors [estrogen, progesterone receptors, and human epidermal growth factor receptor (HER2)], and the treatment approaches frequently include antiestrogen, aromatase inhibitors, and Herceptin. However, the management and prevention strategies due to adverse side effects stress the patients. The unsuccessful treatments cause to raise the drug levels, leading to excessive toxic effects on healthy cells, and the development of multidrug-resistance (MDR) in the tumor cells against chemotherapeutic agents. MDR initially causes the tumor cells to gain a metastatic character, and subsequently, the patients do not respond adequately to treatment. Endoplasmic reticulum (ER) stress is one of the most important mechanisms supporting MDR development. ER stress-mediated chemotherapeutic resistance is very common in aggressive tumors. The in vitro and in vivo experiments on breast tumors indicate that ER stress-activated protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK)- activating transcription factor (ATF4) signal axis plays an important role in the survival of tumors and metastasis. Besides, ER stress-associated oncogenic microRNAs (miRNAs) induce chemoresistance in breast tumors. We aimed to have a look at the development of resistance mechanisms due to ER stress as well as the involvement of ER stress-associated miRNA regulation following the chemotherapeutic regimen in the human breast tumors. We also aimed to draw attention to potential molecular markers and therapeutic targets.
Collapse
Affiliation(s)
- Leila MEHDIZADEHTAPEH
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, İstanbul Kültür University, İstanbulTurkey
| | - Pınar OBAKAN YERLİKAYA
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, İstanbul Kültür University, İstanbulTurkey
| |
Collapse
|
26
|
Bai H, Wang J, Phan CU, Chen Q, Hu X, Shao G, Zhou J, Lai L, Tang G. Cyclodextrin-based host-guest complexes loaded with regorafenib for colorectal cancer treatment. Nat Commun 2021; 12:759. [PMID: 33536421 PMCID: PMC7858623 DOI: 10.1038/s41467-021-21071-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 01/11/2021] [Indexed: 12/11/2022] Open
Abstract
The malignancy of colorectal cancer (CRC) is connected with inflammation and tumor-associated macrophages (TAMs), but effective therapeutics for CRC are limited. To integrate therapeutic targeting with tumor microenvironment (TME) reprogramming, here we develop biocompatible, non-covalent channel-type nanoparticles (CNPs) that are fabricated through host-guest complexation and self-assemble of mannose-modified γ-cyclodextrin (M-γ-CD) with Regorafenib (RG), RG@M-γ-CD CNPs. In addition to its carrier role, M-γ-CD serves as a targeting device and participates in TME regulation. RG@M-γ-CD CNPs attenuate inflammation and inhibit TAM activation by targeting macrophages. They also improve RG's anti-tumor effect by potentiating kinase suppression. In vivo application shows that the channel-type formulation optimizes the pharmacokinetics and bio-distribution of RG. In colitis-associated cancer and CT26 mouse models, RG@M-γ-CD is proven to be a targeted, safe and effective anti-tumor nanomedicine that suppresses tumor cell proliferation, lesions neovascularization, and remodels TME. These findings indicate RG@M-γ-CD CNPs as a potential strategy for CRC treatment.
Collapse
Affiliation(s)
- Hongzhen Bai
- Department of Chemistry, Zhejiang University, 310028, Hangzhou, PR China
| | - Jianwei Wang
- Department of Chemistry, Zhejiang University, 310028, Hangzhou, PR China
| | - Chi Uyen Phan
- Department of Chemistry, Zhejiang University, 310028, Hangzhou, PR China
| | - Qi Chen
- Department of Chemistry, Zhejiang University, 310028, Hangzhou, PR China
| | - Xiurong Hu
- Department of Chemistry, Zhejiang University, 310028, Hangzhou, PR China
| | - Guoqiang Shao
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, 210029, Nanjing, PR China
| | - Jun Zhou
- Department of Chemistry, Zhejiang University, 310028, Hangzhou, PR China
| | - Lihua Lai
- Department of Pharmacology, School of Medicine, Zhejiang University, 310058, Hangzhou, PR China.
| | - Guping Tang
- Department of Chemistry, Zhejiang University, 310028, Hangzhou, PR China.
| |
Collapse
|
27
|
Chen X, Liu Y, Zhang Q, Liu B, Cheng Y, Zhang Y, Sun Y, Liu J, Gen H. Exosomal Long Non-coding RNA HOTTIP Increases Resistance of Colorectal Cancer Cells to Mitomycin via Impairing MiR-214-Mediated Degradation of KPNA3. Front Cell Dev Biol 2021; 8:582723. [PMID: 33585440 PMCID: PMC7876302 DOI: 10.3389/fcell.2020.582723] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 11/12/2020] [Indexed: 12/15/2022] Open
Abstract
It has been reported that long non-coding RNA HOXA distal transcript antisense RNA (lncRNA HOTTIP) functions as a tumor promoter in colorectal cancer (CRC). Hence, we paid attention to exploring whether exosomes could carry lncRNA HOTTIP to affect the mitomycin resistance in CRC and to identify the underlying mechanisms. High expression of HOTTIP was detected in mitomycin-resistant CRC cells. Inhibition of HOTTIP reduced the mitomycin resistance. In the co-culture system of mitomycin-resistant cells or their derived exosomes with CRC cells, the HOTTIP was found to be transferred into the parental cells via extracellular vesicles (EVs) secreted from mitomycin-resistant cells and to contribute to the mitomycin resistance. Based on the bioinformatics databases, possible interaction network of HOTTIP, microRNA-214 (miR-214) and Karyopherin subunit alpha 3 (KPNA3) in CRC was predicted, which was further analyzed by dual-luciferase reporter, RNA binding protein immunoprecipitation and RNA pull-down assays. As HOTTIP down-regulated miR-214 to elevate the KPNA3 expression, HOTTIP enhanced the mitomycin resistance through impairing miR-214-dependent inhibition of KPNA3. Finally, HOTTIP was suggested as an independent factor predicting mitomycin response in patients with CRC. Those data together confirmed the promotive effects of EV-carried HOTTIP on the mitomycin resistance, while targeting HOTTIP might be a promising strategy overcoming drug resistance in CRC.
Collapse
Affiliation(s)
- Xijuan Chen
- Department of Radiation Oncology, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingqiang Liu
- Department of General Surgery, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, China
| | - Qinglan Zhang
- Department of Hematology, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, China
| | - Baoxing Liu
- Department of Chest Surgery, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Cheng
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yonglei Zhang
- Department of General Surgery, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanan Sun
- Department of Radiation Oncology, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, China
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hong Gen
- Department of Radiation Oncology, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
28
|
Li J, Guo W, Bai Y, Deng Y, Yang L, Chen Z, Zhong H, Xu R, Pan H, Shu Y, Yuan Y, Zhou J, Xu N, Liu T, Ma D, Wu C, Cheng Y, Xu J, Chen D, Li W, Sun S, Yu Z, Cao P, Shen L, Chen H, Wang S, Wang H, Fan S, Guo X, Wang N, Han R, Zhang B, Qin S. Safety Profile and Adverse Events of Special Interest for Fruquintinib in Chinese Patients with Previously Treated Metastatic Colorectal Cancer: Analysis of the Phase 3 FRESCO Trial. Adv Ther 2020; 37:4585-4598. [PMID: 32901330 DOI: 10.1007/s12325-020-01477-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Indexed: 10/23/2022]
Abstract
INTRODUCTION In FRESCO (Fruquintinib Efficacy and Safety in 3+ Line Colorectal Cancer Patients) trial, fruquintinib demonstrated a statistically significant and clinically meaningful overall survival benefit in Chinese patients with metastatic colorectal cancer (mCRC). However, its safety profile, including adverse events of special interest (AESIs) and treatment-emergent adverse events (TEAEs) by age, sex, and body mass index (BMI), is not well known. The present analysis evaluated the safety profile and AESIs for fruquintinib in the FRESCO trial. METHODS In FRESCO, eligible Chinese patients were randomized (2:1) to receive fruquintinib (5 mg once daily for 3 weeks, followed by 1 week off in 28-day cycles) or placebo plus best supportive care. Treatment-related AESIs and time to first occurrence of AESIs were summarized. Treatment-related TEAEs by age, sex, and BMI were also summarized. RESULTS A total of 266 patients (95.7%) in the fruquintinib group and 97 (70.8%) in the placebo group had at least one treatment-related TEAE; the mean relative dose intensity was 92% and 98%, respectively. In the fruquintinib group, the most common (in > 40% of patients) treatment-related AESIs were hypertension (55.4%), palmar-plantar erythrodysesthesia syndrome [known as hand-foot skin reaction (HFSR)] (49.3%), and proteinuria (42.1%). The most common treatment-related grade ≥ 3 AESIs (≥ 3% of patients) were hypertension (21.2%), HFSR (10.8%), and proteinuria (3.2%); the median time to onset of these events was 10, 21, and 20 days, respectively. Subgroup analysis by age, sex, and BMI revealed that the frequencies of treatment-related TEAEs were similar across all subgroups, and were consistent with the overall safety profile of fruquintinib. CONCLUSIONS The most common treatment-related grade ≥ 3 AEs were hypertension, HFSR, and proteinuria. The treatment-related TEAE profile of fruquintinib in Chinese patents with mCRC was comparable among different subgroups and consistent with that reported in the overall population. TRIAL REGISTRATION Clinical Trials identifier NCT02314819.
Collapse
|
29
|
Wang WD, Wu GY, Bai KH, Shu LL, Chi PD, He SY, Huang X, Zhang QY, Li L, Wang DW, Dai YJ. A prognostic stemness biomarker CCDC80 reveals acquired drug resistance and immune infiltration in colorectal cancer. Clin Transl Med 2020; 10:e225. [PMID: 33135356 PMCID: PMC7603297 DOI: 10.1002/ctm2.225] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/17/2020] [Accepted: 10/18/2020] [Indexed: 01/05/2023] Open
Affiliation(s)
- Wei-Da Wang
- State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Guo-Yan Wu
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kun-Hao Bai
- State Key Laboratory of Oncology in South China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Endoscopy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ling-Ling Shu
- State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Pei-Dong Chi
- State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Si-Yuan He
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Xin Huang
- State Key Laboratory of Oncology in South China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Qian-Yi Zhang
- State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Liang Li
- State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Da-Wei Wang
- National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-Jun Dai
- State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
30
|
Jiang Y, Yan X, Liu K, Shi Y, Wang C, Hu J, Li Y, Wu Q, Xiang M, Zhao R. Discovering the molecular differences between right- and left-sided colon cancer using machine learning methods. BMC Cancer 2020; 20:1012. [PMID: 33076847 PMCID: PMC7574488 DOI: 10.1186/s12885-020-07507-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 10/07/2020] [Indexed: 12/24/2022] Open
Abstract
Background In recent years, the differences between left-sided colon cancer (LCC) and right-sided colon cancer (RCC) have received increasing attention due to the clinicopathological variation between them. However, some of these differences have remained unclear and conflicting results have been reported. Methods From The Cancer Genome Atlas (TCGA), we obtained RNA sequencing data and gene mutation data on 323 and 283 colon cancer patients, respectively. Differential analysis was firstly done on gene expression data and mutation data between LCC and RCC, separately. Machine learning (ML) methods were then used to select key genes or mutations as features to construct models to classify LCC and RCC patients. Finally, we conducted correlation analysis to identify the correlations between differentially expressed genes (DEGs) and mutations using logistic regression (LR) models. Results We found distinct gene mutation and expression patterns between LCC and RCC patients and further selected the 30 most important mutations and 17 most important gene expression features using ML methods. The classification models created using these features classified LCC and RCC patients with high accuracy (areas under the curve (AUC) of 0.8 and 0.96 for mutation and gene expression data, respectively). The expression of PRAC1 and BRAF V600E mutation (rs113488022) were the most important feature for each model. Correlations of mutations and gene expression data were also identified using LR models. Among them, rs113488022 was found to have significance relevance to the expression of four genes, and thus should be focused on in further study. Conclusions On the basis of ML methods, we found some key molecular differences between LCC and RCC, which could differentiate these two groups of patients with high accuracy. These differences might be key factors behind the variation in clinical features between LCC and RCC and thus help to improve treatment, such as determining the appropriate therapy for patients.
Collapse
Affiliation(s)
- Yimei Jiang
- Department of General Surgery, Ruijin Hospital North, Shanghai Jiaotong University School of Medicine, Shanghai, 201801, China
| | - Xiaowei Yan
- Department of General Surgery, Ruijin Hospital North, Shanghai Jiaotong University School of Medicine, Shanghai, 201801, China
| | - Kun Liu
- Department of General Surgery, Ruijin Hospital North, Shanghai Jiaotong University School of Medicine, Shanghai, 201801, China
| | - Yiqing Shi
- Department of General Surgery, Ruijin Hospital North, Shanghai Jiaotong University School of Medicine, Shanghai, 201801, China
| | - Changgang Wang
- Department of General Surgery, Ruijin Hospital North, Shanghai Jiaotong University School of Medicine, Shanghai, 201801, China
| | - Jiele Hu
- Department of General Surgery, Ruijin Hospital North, Shanghai Jiaotong University School of Medicine, Shanghai, 201801, China
| | - You Li
- Department of General Surgery, Ruijin Hospital North, Shanghai Jiaotong University School of Medicine, Shanghai, 201801, China
| | - Qinghua Wu
- Department of General Surgery, Ruijin Hospital North, Shanghai Jiaotong University School of Medicine, Shanghai, 201801, China
| | - Ming Xiang
- Department of General Surgery, Ruijin Hospital North, Shanghai Jiaotong University School of Medicine, Shanghai, 201801, China.
| | - Ren Zhao
- Department of General Surgery, Ruijin Hospital North, Shanghai Jiaotong University School of Medicine, Shanghai, 201801, China.
| |
Collapse
|
31
|
De Mattia E, Bignucolo A, Toffoli G, Cecchin E. Genetic Markers of the Host to Predict the Efficacy of Colorectal Cancer Targeted Therapy. Curr Med Chem 2020; 27:4249-4273. [PMID: 31298142 DOI: 10.2174/0929867326666190712151417] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 12/19/2018] [Accepted: 01/30/2019] [Indexed: 12/15/2022]
Abstract
The introduction of anti-EGFR (cetuximab and panitumumab) and antiangiogenic (bevacizumab, regorafeninb, ramucirumab, and aflibercept) agents in the therapeutic armamentarium of the metastatic colorectal cancer (CRC) has significantly improved the therapeutic efficacy and patients survival. However, despite the great improvements achieved in the patients life expectation, the high inter-individual heterogeneity in the response to the targeted agents still represent an issue for the management of advanced CRC patients. Even if the role of tumor genetic mutations as predictive markers of drug efficacy has been well-established, the contribution of the host genetic markers is still controversial. Promising results regard the germ-line immune-profile, inflammation and tumor microenvironment. Inherent variations in KRAS 3'UTR region as well as EGF/ EGFR genes were investigated as markers of cetuximab effectiveness. More recently interesting data in the field of anti- EGFR agents were generated also for germ-line variants in genes involved in inflammation (e.g. COX-2, LIFR, IGF1 signaling), immune system (e.g., FCGRs, IL-1RA), and other players of the RAS signaling, including the Hippo pathway related genes (e.g. Rassf, YAP, TAZ). Host genetic variants in VEGF-dependent (i.e., EGF, IGF-1, HIF1α, eNOS, iNOS) and -independent (i.e., EMT cascade, EGFL7) pathways, with specific attention on inflammation and immune system-related factors (e.g., IL-8, CXCR-1/2, CXCR4-CXCL12 axis, TLRs, GADD34, PPP1R15A, ANXA11, MKNK1), were investigated as predictive markers of bevacizumab outcome, generating some promising results. In this review, we aimed to summarize the most recent literature data regarding the potential role of common and rare inhered variants in predicting which CRC patients will benefit more from a specifically targeted drug administration.
Collapse
Affiliation(s)
- Elena De Mattia
- Clinical and Experimental Pharmacology, "Centro di Riferimento Oncologico"- National Cancer Institute, via Franco Gallini 2, 33081, Aviano (PN), Italy
| | - Alessia Bignucolo
- Clinical and Experimental Pharmacology, "Centro di Riferimento Oncologico"- National Cancer Institute, via Franco Gallini 2, 33081, Aviano (PN), Italy
| | - Giuseppe Toffoli
- Clinical and Experimental Pharmacology, "Centro di Riferimento Oncologico"- National Cancer Institute, via Franco Gallini 2, 33081, Aviano (PN), Italy
| | - Erika Cecchin
- Clinical and Experimental Pharmacology, "Centro di Riferimento Oncologico"- National Cancer Institute, via Franco Gallini 2, 33081, Aviano (PN), Italy
| |
Collapse
|
32
|
De Mattia E, Roncato R, Palazzari E, Toffoli G, Cecchin E. Germline and Somatic Pharmacogenomics to Refine Rectal Cancer Patients Selection for Neo-Adjuvant Chemoradiotherapy. Front Pharmacol 2020; 11:897. [PMID: 32625092 PMCID: PMC7311751 DOI: 10.3389/fphar.2020.00897] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/02/2020] [Indexed: 12/25/2022] Open
Abstract
Neoadjuvant chemoradiotherapy (nCRT) followed by radical surgery is the standard of care for patients with Locally Advanced Rectal Cancer (LARC). Current selection for nCRT is based on clinical criteria regardless of any molecular marker. Pharmacogenomics may be a useful strategy to personalize and optimize nCRT in LARC. This review aims to summarize the most recent and relevant findings about the role of germline and somatic pharmacogenomics in the prediction of nCRT outcome in patients with LARC, discussing the state of the art of their application in the clinical practice. A systematic literature search of the PubMed database was completed to identify relevant English-language papers published up to January 2020. The chemotherapeutic backbone of nCRT is represented by fluoropyrimidines, mainly metabolized by DPD (Dihydro-Pyrimidine Dehydrogenase, DPYD). The clinical impact of testing DPYD*2A, DPYD*13, c.2846A > T and c.1236G > A-HapB3 before a fluoropyrimidines administration to increase treatment safety is widely acknowledged. Other relevant target genes are TYMS (Thymidylate Synthase) and MTHFR (Methylene-Tetrahydro-Folate Reductase), whose polymorphisms were mainly studied as potential markers of treatment efficacy in LARC. A pivotal role of a TYMS polymorphism in the gene promoter region (rs34743033) was reported and was pioneeringly used to guide nCRT treatment in a phase II study. The pharmacogenomic analysis of other pathways mostly involved in the cellular response to radiation damage, as the DNA repair and the activation of the inflammatory cascade, provided less consistent results. A high rate of somatic mutation in genes belonging to PI3K (Phosphatidyl-Inositol 3-Kinase) and MAPK (Mitogen-Activated Protein Kinase) pathways, as BRAF (V-raf murine sarcoma viral oncogene homolog B1), KRAS (Kirsten Rat Sarcoma viral oncogene homolog), NRAS (Neuroblastoma RAS viral (v-ras) oncogene homolog), PIK3CA (Phosphatidyl-Inositol-4,5-bisphosphate 3-Kinase, Catalytic Subunit Alpha), as well as TP53 (Tumor Protein 53) was reported in LARC. Their pharmacogenomic role, already defined in colorectal cancer, is under investigation in LARC with promising results concerning specific somatic mutations in KRAS and TP53, as predictors of tumor response and prognosis. The availability of circulating tumor DNA in plasma may also represent an opportunity to monitor somatic mutations in course of therapy.
Collapse
Affiliation(s)
- Elena De Mattia
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Rossana Roncato
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Elisa Palazzari
- Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Giuseppe Toffoli
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Erika Cecchin
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| |
Collapse
|
33
|
Meng X, Xu Y, Lu Q, Sun L, An X, Zhang J, Chen J, Gao Y, Zhang Y, Ning X. Ultrasound-responsive alkaline nanorobots for the treatment of lactic acidosis-mediated doxorubicin resistance. NANOSCALE 2020; 12:13801-13810. [PMID: 32573588 DOI: 10.1039/d0nr03726e] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Lactic acidosis is one of the key characteristics of the tumor microenvironment (TME), and plays a critical role in therapy resistance, making it an attractive target for enhancing anticancer treatment. However, no effective systems exhibit the ability to selectively neutralize tumor lactic acidosis in a controlled manner. Here, we have developed novel ultrasound-responsive alkaline nanorobots (AN-DSP), composed of PLGA nanoparticles containing doxorubicin (DOX), sodium carbonate (Na2CO3) and perfluorocarbon (PFC), for recovering from lactic acidosis-mediated drug resistance. AN-DSP exhibit sensitive response to ultrasound stimulation, and rapidly release Na2CO3 to neutralize lactic acidosis, consequently enhancing DOX susceptibility in vitro and in vivo. Particularly, our nanorobots autonomously accumulate in tumors by an enhanced permeability and retention effect, and can specifically disrupt the tumor acidic microenvironment in response to external ultrasonic powering, resulting in the inhibition of tumor growth with minimal adverse effects. Therefore, AN-DSP represent a promising approach for selectively overcoming tumor lactic acidosis induced therapeutic resistance.
Collapse
Affiliation(s)
- Xia Meng
- National Laboratory of Solid State Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, 210093, Nanjing, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Duran G, Cruz R, Simoes AR, Barros F, Giráldez JM, Bernárdez B, Anido U, Candamio S, López-López R, Carracedo Á, Lamas MJ. Efficacy and toxicity of adjuvant chemotherapy on colorectal cancer patients: how much influence from the genetics? J Chemother 2020; 32:310-322. [PMID: 32441565 DOI: 10.1080/1120009x.2020.1764281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We studied the predictive value for response and toxicity of functional polymorphisms in genes involved in the oxaliplatin/fluorouracil pathway in colorectal cancer patients. One hundred and twenty-seven (127) patients were treated with curative intended surgery followed by adjuvant chemotherapy with FOLFOX (fluorouracil, leucovorin and oxaliplatin) regimen. The median age was 65.53 (27-80) years (66.9% male, 59.1% rectum). The median follow-up was 8.5 years (IQR, 4.1-9.4). At the end of follow-up, 59 patients (46.5%) had relapsed or died in the whole study population. We did find that XRCC1GG genotype is associated with a higher risk of developing haematologic toxicity. Furthermore, we report a significant association of the TS 3'UTR 6 bp/6 bp polymorphism and the XRCC1 rs25487 with a higher risk of developing anaemia and diarrhoea, respectively. On the other hand, none of the studied polymorphisms showed clinically relevant association with disease-free survival and overall survival or early failure to adjuvant FOLFOX therapy.
Collapse
Affiliation(s)
- Goretti Duran
- Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Pharmacy Department, University Hospital of Santiago (SERGAS), Santiago de Compostela, Spain
| | - Raquel Cruz
- Center for Biomedical Research on Rare Diseases (CIBERER), Genomics Medicine Group, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ana Rita Simoes
- Fundación Instituto de Investigación Sanitaria de Santiago (FIDIS), Santiago de Compostela, Spain.,Departamento de Ciencias Forenses, Anatomía Patolóxica, Xinecoloxía, Obstetricia e Pediatría, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain.,Genomics Medicine Group, Galician Public Foundation of Genomic Medicine (FPGMX), Santiago de Compostela, Spain
| | - Francisco Barros
- Genomics Medicine Group, Galician Public Foundation of Genomic Medicine (FPGMX), Santiago de Compostela, Spain
| | - José María Giráldez
- Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Pharmacy Department, University Hospital of Santiago (SERGAS), Santiago de Compostela, Spain
| | - Beatriz Bernárdez
- Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Pharmacy Department, University Hospital of Santiago (SERGAS), Santiago de Compostela, Spain
| | - Urbano Anido
- Translational Medical Oncology, Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain.,Medical Oncology Department, University Hospital of Santiago (SERGAS), Santiago de Compostela, Spain
| | - Sonia Candamio
- Translational Medical Oncology, Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain.,Medical Oncology Department, University Hospital of Santiago (SERGAS), Santiago de Compostela, Spain
| | - Rafael López-López
- Translational Medical Oncology, Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain.,Medical Oncology Department, University Hospital of Santiago (SERGAS), Santiago de Compostela, Spain.,Liquid Biopsy Analysis Unit, Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain
| | - Ángel Carracedo
- Genomics Medicine Group, CIBERER, Galician Public Foundation of Genomic Medicine (FPGMX), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - María Jesús Lamas
- Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Pharmacy Department, University Hospital of Santiago (SERGAS), Santiago de Compostela, Spain
| |
Collapse
|
35
|
Dai DP, Yu LZ, Ye MF. LincRNA-p21 reverses irinotecan resistance in colon cancer cells via the PI3K/AKT signaling pathway. Shijie Huaren Xiaohua Zazhi 2019; 27:1356-1364. [DOI: 10.11569/wcjd.v27.i22.1356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Irinotecan (camptothecin-11, CPT-11) is a first-line chemotherapy drug for advanced colon cancer, but CPT-11 resistance limits its efficacy. Studying the mechanism of CPT-11 resistance in colon cancer and restoring the sensitivity of colon cancer cells to CPT-11 are of great clinical value in prolonging the life time of colon cancer patients.
AIM To investigate the effect and mechanism of long intergenic non-coding RNA-p21 (lincRNA-p21) on CPT-11 resistance in colon cancer cells.
METHODS HCT-8 and SW480 cells were used to construct irinotecan-resistant HCT-8/CPT-11 and SW480/CPT-11 cell lines by continuously exposing them to increasing concentrations of CPT-11, and the expression of lincRNA-p21 in the cells was detected by real-time quantitative polymerase chain reaction (RT-qPCR). After transfection with pcDNA-lincRNA-p21 or si-lincRNA-p21, the effect of CPT-11 on the viability of HCT-8/CPT-11 cells or SW480/CPT-11 cells was measured by cell counting kit-8 (CCK-8) assay. The regulatory effect of lincRNA-p21 on the phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) pathway was preliminarily analyzed by Western blot. After pretreatment with PI3K/AKT pathway inhibitor LY294002 prior to transfection with si-lincRNA-p21, or pretreatment with PI3K/AKT pathway agonist Recilisib prior to transfection with pcDNA-lincRNA-p21, the effect of CPT-11 on cell viability in HCT-8/CPT-11 cells or SW480/CPT-11 cells was measured by CCK-8 assay.
RESULTS LincRNA-p21 expression in CPT-11 resistant cells was significantly lower than that in parental cells. Overexpression of lincRNA-p21 inhibited the resistance of HCT-8/CPT-11 cells and SW480/CPT-11 cells to CPT-11, while knockdown of lincRNA-p21 enhanced the resistance of HCT-8/CPT-11 cells and SW480/CPT-11 cells to CPT-11. Western blot results showed that overexpression of lincRNA-p21 inhibited the activity of the PI3K/AKT pathway, while knockdown of lincRNA-p21 enhanced the activity of the PI3K/AKT pathway. LY294002 inhibited the promotive effect of lincRNA-p21 knockdown on CPT-11 resistance, while Recilisib inhibited the inhibitive effect of lincRNA-p21 overexpression on CPT-11 resistance.
CONCLUSION Up-regulation of lincRNA-p21 can inhibit the CPT-11 resistance of colorectal cancer cells, while down-regulation of lincRNA-p21 can promote their CPT-11 resistance, which may be related to the regulation of the PI3K/AKT signaling activity by lincRNA-p21.
Collapse
Affiliation(s)
- Dan-Ping Dai
- Department of Pharmacy, Taizhou Hospital, Taizhou Grace Medical Center (Group), Taizhou 317000, Zhejiang Province, China
| | - Ling-Zhi Yu
- Department of Pharmacy, Taizhou Hospital, Taizhou Grace Medical Center (Group), Taizhou 317000, Zhejiang Province, China
| | - Meng-Fei Ye
- Department of Pharmacy, Taizhou Hospital, Taizhou Grace Medical Center (Group), Taizhou 317000, Zhejiang Province, China
| |
Collapse
|
36
|
De Mattia E, Polesel J, Roncato R, Labriet A, Bignucolo A, Dreussi E, Romanato L, Guardascione M, Buonadonna A, D'Andrea M, Lévesque E, Jonker D, Couture F, Guillemette C, Cecchin E, Toffoli G. Germline Polymorphisms in the Nuclear Receptors PXR and VDR as Novel Prognostic Markers in Metastatic Colorectal Cancer Patients Treated With FOLFIRI. Front Oncol 2019; 9:1312. [PMID: 31850208 PMCID: PMC6901926 DOI: 10.3389/fonc.2019.01312] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 11/11/2019] [Indexed: 12/16/2022] Open
Abstract
Nuclear receptors act as mediators of cancer-related inflammation and gene expression. They have a regulatory effect on genes encoding proteins related to drug adsorption, distribution, metabolism, and excretion. The aim of the present study was to highlight novel prognostic markers among polymorphisms in genes encoding for nuclear receptor proteins and inflammation-related cytokines in patients treated with a FOLFIRI regimen. This study included two independent cohorts comprising a total of 337 mCRC patients homogeneously treated with first-line FOLFIRI. Genotyping of 246 haplotype-tagging polymorphisms in 22 genes was performed using bead array technology. The NR1I2 (PXR)-rs1054190 and VDR-rs7299460 polymorphisms were significantly associated with patient overall survival (OS). A detrimental effect of the NR1I2 rs1054190-TT genotype on OS was observed in both the discovery and replication cohorts (HR = 6.84, P = 0.0021, q-value = 0.1278 and HR = 3.56, P = 0.0414, respectively). Patients harboring the NR1I2 rs1054190-TT genotype had a median OS of 9 months vs. 21 months in patients with C-allele (P < 0.0001 log-rank test). VDR rs7299460-T was consistently associated with a longer OS in both cohorts (discovery: HR = 0.61, P = 0.0075, q-value = 0.1535; replication: HR = 0.57, P = 0.0477). Patients with the VDR rs7299460-T allele had a median OS of 23 months compared to 18 months in those with the CC genotype (P = 0.0489, log-rank test). The NR1I2-rs1054190 polymorphism also had an effect on the duration of progression-free survival, consistent with the effect observed on OS. Two novel prognostic markers for mCRC treated with FOLFIRI were described and, if validated by prospective trials, have a potential application in the management of these patients.
Collapse
Affiliation(s)
- Elena De Mattia
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Jerry Polesel
- Unit of Cancer Epidemiology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Rossana Roncato
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Adrien Labriet
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec (CHU de Québec) Research Center and Faculty of Pharmacy, Laval University, Quebec City, QC, Canada
| | - Alessia Bignucolo
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Eva Dreussi
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Loredana Romanato
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Michela Guardascione
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Angela Buonadonna
- Medical Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Mario D'Andrea
- Medical Oncology Unit, "San Filippo Neri Hospital", Rome, Italy
| | - Eric Lévesque
- CHU de Québec Research Center and Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Derek Jonker
- Division of Medical Oncology, Department of Medicine, Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| | - Félix Couture
- CHU de Québec Research Center and Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Chantal Guillemette
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec (CHU de Québec) Research Center and Faculty of Pharmacy, Laval University, Quebec City, QC, Canada
| | - Erika Cecchin
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Giuseppe Toffoli
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| |
Collapse
|
37
|
Lee HW, Son E, Lee K, Lee Y, Kim Y, Lee JC, Lim Y, Hur M, Kim D, Nam DH. Promising Therapeutic Efficacy of GC1118, an Anti-EGFR Antibody, against KRAS Mutation-Driven Colorectal Cancer Patient-Derived Xenografts. Int J Mol Sci 2019; 20:ijms20235894. [PMID: 31771279 PMCID: PMC6928876 DOI: 10.3390/ijms20235894] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 11/22/2019] [Indexed: 02/07/2023] Open
Abstract
Epidermal growth factor receptor (EGFR)-targeted monoclonal antibodies, including cetuximab and panitumumab, are used to treat metastatic colorectal cancer (mCRC). However, this treatment is only effective for a small subset of mCRC patients positive for the wild-type KRAS GTPase. GC1118 is a novel, fully humanized anti-EGFR IgG1 antibody that displays potent inhibitory effects on high-affinity EGFR ligand-induced signaling and enhanced antibody-mediated cytotoxicity. In this study, using 51 CRC patient-derived xenografts (PDXs), we showed that KRAS mutants expressed remarkably elevated autocrine levels of high-affinity EGFR ligands compared with wild-type KRAS. In three KRAS-mutant CRCPDXs, GC1118 was more effective than cetuximab, whereas the two agents demonstrated comparable efficacy against three wild-type KRAS PDXs. Persistent phosphatidylinositol-3-kinase (PI3K)/AKT signaling was thought to underlie resistance to GC1118. In support of these findings, a preliminary improved anti-cancer response was observed in a CRC PDX harboring mutated KRAS with intrinsically high AKT activity using GC1118 combined with the dual PI3K/mammalian target of rapamycin (mTOR)/AKT inhibitor BEZ-235, without observed toxicity. Taken together, the superior antitumor efficacy of GC1118 alone or in combination with PI3K/mTOR/AKT inhibitors shows great therapeutic potential for the treatment of KRAS-mutant mCRC with elevated ratios of high- to low-affinity EGFR ligands and PI3K-AKT pathway activation.
Collapse
Affiliation(s)
- Hye Won Lee
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16149, Korea;
- Single Cell Network Research Center, Sungkyunkwan University, Suwon 16149, Korea
| | - Eunju Son
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul 06351, Korea; (E.S.); (K.L.); (Y.L.); (Y.K.)
| | - Kyoungmin Lee
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul 06351, Korea; (E.S.); (K.L.); (Y.L.); (Y.K.)
- Department of Health Science and Technology, Samsung Advanced Institute for Health Science and Technology, Sungkyunkwan University, Seoul 06351, Korea
| | - Yeri Lee
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul 06351, Korea; (E.S.); (K.L.); (Y.L.); (Y.K.)
- Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea
| | - Yejin Kim
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul 06351, Korea; (E.S.); (K.L.); (Y.L.); (Y.K.)
- Department of Health Science and Technology, Samsung Advanced Institute for Health Science and Technology, Sungkyunkwan University, Seoul 06351, Korea
| | - Jae-Chul Lee
- Translational Research 1 Team, MOGAM Institute for Biomedical Research, Yongin 16924, Korea; (J.-C.L.); (Y.L.); (M.H.)
| | - Yangmi Lim
- Translational Research 1 Team, MOGAM Institute for Biomedical Research, Yongin 16924, Korea; (J.-C.L.); (Y.L.); (M.H.)
| | - Minkyu Hur
- Translational Research 1 Team, MOGAM Institute for Biomedical Research, Yongin 16924, Korea; (J.-C.L.); (Y.L.); (M.H.)
| | - Donggeon Kim
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul 06351, Korea; (E.S.); (K.L.); (Y.L.); (Y.K.)
- Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea
- Correspondence: (D.K.); (D.-H.N.); Tel.: +82-02-2148-7723 (D.K.); +82-02-3410-3497 (D.-H.N.)
| | - Do-Hyun Nam
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul 06351, Korea; (E.S.); (K.L.); (Y.L.); (Y.K.)
- Department of Health Science and Technology, Samsung Advanced Institute for Health Science and Technology, Sungkyunkwan University, Seoul 06351, Korea
- Department of Neurosurgery, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul 06531, Korea
- Correspondence: (D.K.); (D.-H.N.); Tel.: +82-02-2148-7723 (D.K.); +82-02-3410-3497 (D.-H.N.)
| |
Collapse
|
38
|
Xu X, Chen X, Xu M, Liu X, Pan B, Qin J, Xu T, Zeng K, Pan Y, He B, Sun H, Sun L, Wang S. miR-375-3p suppresses tumorigenesis and partially reverses chemoresistance by targeting YAP1 and SP1 in colorectal cancer cells. Aging (Albany NY) 2019; 11:7357-7385. [PMID: 31543507 PMCID: PMC6781994 DOI: 10.18632/aging.102214] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 08/13/2019] [Indexed: 02/07/2023]
Abstract
Clinically, one of the principal factors in the failure of advanced colorectal cancer (CRC) treatment is chemoresistance to 5-fluorouracil (5FU)-based chemotherapy. Although microRNA-375-3p (miR-375) is considered a tumor suppressor in multiple cancers, the mechanism of miR-375 in the regulation of drug resistance in CRC remains unclear. In this study, we investigated the chemosensitivity of miR-375 to 5FU in CRC from biological and clinical aspects. We found that miR-375 was significantly downregulated in CRC tissues and cell lines, and low miR-375 expression was strongly correlated with poor overall survival in CRC patients. Overexpression of miR-375 sensitized CRC cells to a broad spectrum of chemotherapeutic drugs in vitro and in vivo. Further mechanistic analysis demonstrated that miR-375 enhanced CRC cell sensitivity to 5FU by directly targeting YAP1 and SP1. MiR-375 downregulated YAP1, resulting in reduced expression of the Hippo-YAP1 pathway downstream genes CTGF, cyclin D1 and BIRC5 (also known as survivin). Overall, miR-375 was confirmed as a prospective molecular biomarker in the chemoresistance and prognosis of CRC patients, and the synergy between miR-375 and chemotherapeutic drugs could be a promising therapeutic strategy for CRC patients, especially with chemoresistance.
Collapse
Affiliation(s)
- Xueni Xu
- School of Medicine, Southeast University, Nanjing 210009, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xiaoxiang Chen
- School of Medicine, Southeast University, Nanjing 210009, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Mu Xu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xiangxiang Liu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Bei Pan
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Jian Qin
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Tao Xu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Kaixuan Zeng
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Yuqin Pan
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Bangshun He
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Huiling Sun
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Li Sun
- Department of Laboratory Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Shukui Wang
- School of Medicine, Southeast University, Nanjing 210009, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| |
Collapse
|
39
|
Assaraf YG, Brozovic A, Gonçalves AC, Jurkovicova D, Linē A, Machuqueiro M, Saponara S, Sarmento-Ribeiro AB, Xavier CP, Vasconcelos MH. The multi-factorial nature of clinical multidrug resistance in cancer. Drug Resist Updat 2019; 46:100645. [PMID: 31585396 DOI: 10.1016/j.drup.2019.100645] [Citation(s) in RCA: 196] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/05/2019] [Accepted: 09/14/2019] [Indexed: 12/16/2022]
|
40
|
Chen Z, Jiang L. The clinical application of fruquintinib on colorectal cancer. Expert Rev Clin Pharmacol 2019; 12:713-721. [PMID: 31177854 DOI: 10.1080/17512433.2019.1630272] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Zhongguang Chen
- Department of Pharmaceutical, Central Hospital of Linyi City, Yishui, Shandong, China
| | - Lili Jiang
- Ultrasound Medical Department, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
41
|
De Mattia E, Roncato R, Dalle Fratte C, Ecca F, Toffoli G, Cecchin E. The use of pharmacogenetics to increase the safety of colorectal cancer patients treated with fluoropyrimidines. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:116-130. [PMID: 35582139 PMCID: PMC9019179 DOI: 10.20517/cdr.2019.04] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/31/2019] [Accepted: 02/15/2019] [Indexed: 06/02/2023]
Abstract
Fluoropyrimidines (FP) are given in the combination treatment of the advanced disease or as monotherapy in the neo-adjuvant and adjuvant treatment of colorectal cancerand other solid tumors including breast, head and neck and gastric cancer. FP present a narrow therapeutic index with 10 to 26% of patients experiencing acute severe or life-threatening toxicity. With the high number of patients receiving FP-based therapies, and the significant effects of toxicities on their quality of life, the prevention of FP-related adverse events is of major clinical interest. Host genetic variants in the rate limiting enzyme dihydropyrimidine dehydrogenase (DPYD) gene are related to the occurrence of extremely severe, early onset toxicity in FP treated patients. The pre-treatment diagnostic test of 4 DPYD genetic polymorphisms is suggested by the currently available pharmacogenetic guidelines. Several prospective implementation projects are ongoing to support the introduction of up-front genotyping of the patients in clinical practice. Multiple pharmacogenetic studies tried to assess the predictive role of other polymorphisms in genes involved in the FP pharmacokinetics/pharmacodynamic pathways, TYMS and MTHFR, but no additional clinically validated genetic markers of toxicity are available to date. The development of next-generation sequencing platforms opens new possibilities to highlight previously unreported genetic markers. Moreover, the investigation of the genetic variation in the patients immunological system, a pivotal target in cancer treatment, could bring notable advances in the field. This review will describe the most recent literature on the use of pharmacogenetics to increase the safety of a treatment based on FP administration in colorectal cancer patients.
Collapse
Affiliation(s)
- Elena De Mattia
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano 33081, Italy
| | - Rossana Roncato
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano 33081, Italy
| | - Chiara Dalle Fratte
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano 33081, Italy
| | - Fabrizio Ecca
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano 33081, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano 33081, Italy
| | - Erika Cecchin
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano 33081, Italy
| |
Collapse
|
42
|
Skarkova V, Kralova V, Vitovcova B, Rudolf E. Selected Aspects of Chemoresistance Mechanisms in Colorectal Carcinoma-A Focus on Epithelial-to-Mesenchymal Transition, Autophagy, and Apoptosis. Cells 2019; 8:cells8030234. [PMID: 30871055 PMCID: PMC6468859 DOI: 10.3390/cells8030234] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/08/2019] [Accepted: 03/08/2019] [Indexed: 12/15/2022] Open
Abstract
Chemoresistance has been found in all malignant tumors including colorectal carcinoma (CRC). Nowadays chemoresistance is understood as a major reason for therapy failure, with consequent tumor growth and spreading leading ultimately to the patient's premature death. The chemotherapy-related resistance of malignant colonocytes may be manifested in diverse mechanisms that may exist both prior to the onset of the therapy or after it. The ultimate function of this chemoresistance is to ensure the survival of malignant cells through continuing adaptation within an organism, therefore, the nature and spectrum of cell-survival strategies in CRC represent a highly significant target of scientific inquiry. Among these survival strategies employed by CRC cells, three unique but significantly linked phenomena stand out-epithelial-to-mesenchymal transition (EMT), autophagy, and cell death. In this mini-review, current knowledge concerning all three mechanisms including their emergence, timeline, regulation, and mutual relationships will be presented and discussed.
Collapse
Affiliation(s)
- Veronika Skarkova
- Department of Medical Biology and Genetics, Charles University, Faculty of Medicine in Hradec Králové, Zborovská 2089, 500 03 Hradec Králové, Czech Republic.
| | - Vera Kralova
- Department of Medical Biology and Genetics, Charles University, Faculty of Medicine in Hradec Králové, Zborovská 2089, 500 03 Hradec Králové, Czech Republic.
| | - Barbora Vitovcova
- Department of Medical Biology and Genetics, Charles University, Faculty of Medicine in Hradec Králové, Zborovská 2089, 500 03 Hradec Králové, Czech Republic.
| | - Emil Rudolf
- Department of Medical Biology and Genetics, Charles University, Faculty of Medicine in Hradec Králové, Zborovská 2089, 500 03 Hradec Králové, Czech Republic.
| |
Collapse
|
43
|
Jiraskova K, Hughes DJ, Brezina S, Gumpenberger T, Veskrnova V, Buchler T, Schneiderova M, Levy M, Liska V, Vodenkova S, Di Gaetano C, Naccarati A, Pardini B, Vymetalkova V, Gsur A, Vodicka P. Functional Polymorphisms in DNA Repair Genes Are Associated with Sporadic Colorectal Cancer Susceptibility and Clinical Outcome. Int J Mol Sci 2018; 20:E97. [PMID: 30591675 PMCID: PMC6337670 DOI: 10.3390/ijms20010097] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/18/2018] [Accepted: 12/21/2018] [Indexed: 02/06/2023] Open
Abstract
DNA repair processes are involved in both the onset and treatment efficacy of colorectal cancer (CRC). A change of a single nucleotide causing an amino acid substitution in the corresponding protein may alter the efficiency of DNA repair, thus modifying the CRC susceptibility and clinical outcome. We performed a candidate gene approach in order to analyze the association of non-synonymous single nucleotide polymorphisms (nsSNPs) in the genes covering the main DNA repair pathways with CRC risk and clinical outcome modifications. Our candidate polymorphisms were selected according to the foremost genomic and functional prediction databases. Sixteen nsSNPs in 12 DNA repair genes were evaluated in cohorts from the Czech Republic and Austria. Apart from the tumor-node-metastasis (TNM) stage, which occurred as the main prognostic factor in all of the performed analyses, we observed several significant associations of different nsSNPs with survival and clinical outcomes in both cohorts. However, only some of the genes (REV3L, POLQ, and NEIL3) were prominently defined as prediction factors in the classification and regression tree analysis; therefore, the study suggests their association for patient survival. In summary, we provide observational and bioinformatics evidence that even subtle alterations in specific proteins of the DNA repair pathways may contribute to CRC susceptibility and clinical outcome.
Collapse
Affiliation(s)
- Katerina Jiraskova
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 128 00 Prague, Czech Republic.
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague, Czech Republic.
| | - David J Hughes
- Cancer Biology and Therapeutics Group, UCD Conway Institute, University College Dublin, Dublin 4, Ireland.
| | - Stefanie Brezina
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria.
| | - Tanja Gumpenberger
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria.
| | - Veronika Veskrnova
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer Hospital, Videnska 800, 140 59 Prague, Czech Republic.
| | - Tomas Buchler
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer Hospital, Videnska 800, 140 59 Prague, Czech Republic.
| | - Michaela Schneiderova
- Department of Surgery, General University Hospital in Prague, U Nemocnice 499/2, 128 08 Prague, Czech Republic.
| | - Miroslav Levy
- Department of Surgery, First Faculty of Medicine, Charles University and Thomayer Hospital, Thomayerova 815/5, 140 00 Prague, Czech Republic.
| | - Vaclav Liska
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University in Prague, 323 00 Pilsen, Czech Republic.
- Department of Surgery, Medical School in Pilsen, Charles University, Alej svobody 80, 304 600 Pilsen, Czech Republic.
| | - Sona Vodenkova
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 128 00 Prague, Czech Republic.
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague, Czech Republic.
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Ruska 2411/87, 100 00 Prague, Czech Republic.
| | - Cornelia Di Gaetano
- Molecular and Genetic Epidemiology; Genomic Variation in Human Populations and Complex Diseases, IIGM Italian Institute for Genomic Medicine, Via Nizza 52, 10126 Turin, Italy.
- Department of Medical Sciences, University of Turin, Corso Dogliotti 14, 10126 Turin, Italy.
| | - Alessio Naccarati
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague, Czech Republic.
- Molecular and Genetic Epidemiology; Genomic Variation in Human Populations and Complex Diseases, IIGM Italian Institute for Genomic Medicine, Via Nizza 52, 10126 Turin, Italy.
| | - Barbara Pardini
- Molecular and Genetic Epidemiology; Genomic Variation in Human Populations and Complex Diseases, IIGM Italian Institute for Genomic Medicine, Via Nizza 52, 10126 Turin, Italy.
- Department of Medical Sciences, University of Turin, Corso Dogliotti 14, 10126 Turin, Italy.
| | - Veronika Vymetalkova
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 128 00 Prague, Czech Republic.
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague, Czech Republic.
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University in Prague, 323 00 Pilsen, Czech Republic.
| | - Andrea Gsur
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria.
| | - Pavel Vodicka
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 128 00 Prague, Czech Republic.
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague, Czech Republic.
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University in Prague, 323 00 Pilsen, Czech Republic.
| |
Collapse
|
44
|
Yu KH, Ricigliano M, McCarthy B, Chou JF, Capanu M, Cooper B, Bartlett A, Covington C, Lowery MA, O'Reilly EM. Circulating Tumor and Invasive Cell Gene Expression Profile Predicts Treatment Response and Survival in Pancreatic Adenocarcinoma. Cancers (Basel) 2018; 10:cancers10120467. [PMID: 30477242 PMCID: PMC6315371 DOI: 10.3390/cancers10120467] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/17/2018] [Accepted: 11/16/2018] [Indexed: 12/24/2022] Open
Abstract
Previous studies have shown that pharmacogenomic modeling of circulating tumor and invasive cells (CTICs) can predict response of pancreatic ductal adenocarcinoma (PDAC) to combination chemotherapy, predominantly 5-fluorouracil-based. We hypothesized that a similar approach could be developed to predict treatment response to standard frontline gemcitabine with nab-paclitaxel (G/nab-P) chemotherapy. Gene expression profiles for responsiveness to G/nab-P were determined in cell lines and a test set of patient samples. A prospective clinical trial was conducted, enrolling 37 patients with advanced PDAC who received G/nab-P. Peripheral blood was collected prior to treatment, after two months of treatment, and at progression. The CTICs were isolated based on a phenotype of collagen invasion. The RNA was isolated, cDNA synthesized, and qPCR gene expression analyzed. Patients were most closely matched to one of three chemotherapy response templates. Circulating tumor and invasive cells' SMAD4 expression was measured serially. The CTICs were reliably isolated and profiled from peripheral blood prior to and during chemotherapy treatment. Individual patients could be matched to distinct response templates predicting differential responses to G/nab-P treatment. Progression free survival was significantly correlated to response prediction and ΔSMAD4 was significantly associated with disease progression. These findings support phenotypic profiling and ΔSMAD4 of CTICs as promising clinical tools for choosing effective therapy in advanced PDAC, and for anticipating disease progression.
Collapse
Affiliation(s)
- Kenneth H Yu
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
- Weill Cornell Medical College, New York, NY 10065, USA.
| | | | | | - Joanne F Chou
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
- Weill Cornell Medical College, New York, NY 10065, USA.
| | - Marinela Capanu
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
- Weill Cornell Medical College, New York, NY 10065, USA.
| | | | | | | | - Maeve A Lowery
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Eileen M O'Reilly
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
- Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
45
|
De Mattia E, Dreussi E, Montico M, Gagno S, Zanusso C, Quartuccio L, De Vita S, Guardascione M, Buonadonna A, D'Andrea M, Pella N, Favaretto A, Mini E, Nobili S, Romanato L, Cecchin E, Toffoli G. A Clinical-Genetic Score to Identify Surgically Resected Colorectal Cancer Patients Benefiting From an Adjuvant Fluoropyrimidine-Based Therapy. Front Pharmacol 2018; 9:1101. [PMID: 30337874 PMCID: PMC6180157 DOI: 10.3389/fphar.2018.01101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/10/2018] [Indexed: 12/20/2022] Open
Abstract
There are clinical challenges related to adjuvant treatment in colorectal cancer (CRC) and novel molecular markers are needed for better risk stratification of patients. Our aim was to integrate our previously reported clinical-genetic prognostic score with new immunogenetic markers of 5-year disease-free survival (DFS) to evaluate the recurrence risk stratification before fluoropyrimidine (FL)-based adjuvant therapy. The study population included a total of 270 stage II-III CRC patients treated with adjuvant FL with (FL + OXA, n = 119) or without oxaliplatin (FL, n = 151). Patients were genotyped for a panel of 192 tagging polymorphisms in 34 immune-related genes. The IFNG-rs1861494 polymorphism was associated with worse DFS in the FL + OXA (HR = 2.14, 95%CI 1.13–4.08; P = 0.020, q-value = 0.249) and FL (HR = 1.97, 95%CI 1.00–3.86; P = 0.049) cohorts, according to a dominant model. The integration of IFNG-rs1861494 in our previous clinical genetic multiparametric score of DFS improved the patients’ risk stratification (Log-rank P = 0.0026 in the pooled population). These findings could improve the discrimination of patients who would benefit from adjuvant treatment. In addition, the results may help better elucidate the interplay between the immune system and chemotherapeutics and help determine the efficacy of anti-tumor strategies.
Collapse
Affiliation(s)
- Elena De Mattia
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Eva Dreussi
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Marcella Montico
- Scientific Directorate, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Sara Gagno
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Chiara Zanusso
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Luca Quartuccio
- Department of Medical Area (DAME), Rheumatology Clinic, Santa Maria della Misericordia University Hospital, Udine, Italy
| | - Salvatore De Vita
- Department of Medical Area (DAME), Rheumatology Clinic, Santa Maria della Misericordia University Hospital, Udine, Italy
| | - Michela Guardascione
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Angela Buonadonna
- Medical Oncology Unit B, CRO Aviano National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Mario D'Andrea
- Medical Oncology Unit, "San Filippo Neri Hospital", Rome, Italy
| | | | | | - Enrico Mini
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Stefania Nobili
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Loredana Romanato
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Erika Cecchin
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| |
Collapse
|
46
|
Cecchin E, De Mattia E, Ecca F, Toffoli G. Host genetic profiling to increase drug safety in colorectal cancer from discovery to implementation. Drug Resist Updat 2018; 39:18-40. [PMID: 30075835 DOI: 10.1016/j.drup.2018.07.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/11/2018] [Accepted: 07/06/2018] [Indexed: 02/07/2023]
Abstract
Adverse events affect the pharmacological treatment of approximately 90% of colorectal cancer (CRC) patients at any stage of the disease. Chemotherapy including fluoropyrimidines, irinotecan, and oxaliplatin is the cornerstone of the pharmacological treatment of CRC. The introduction of novel targeted agents, as anti-EGFR (i.e. cetuximab, panitumumab) and antiangiogenic (i.e. bevacizumab, ziv-aflibercept, regorafenib, and ramucirumab) molecules, into the oncologist's toolbox has led to significant improvements in the life expectancy of advanced CRC patients, but with a substantial increase in toxicity burden. In this respect, pharmacogenomics has largely been applied to the personalization of CRC chemotherapy, focusing mainly on the study of inhered polymorphisms in genes encoding phase I and II enzymes, ATP-binding cassette (ABC)/solute carrier (SLC) membrane transporters, proteins involved in DNA repair, folate pathway and immune response. These research efforts have led to the identification of some validated genetic markers of chemotherapy toxicity, for fluoropyrimidines and irinotecan. No validated genetic determinants of oxaliplatin-specific toxicity, as peripheral neuropathy, has thus far been established. The contribution of host genetic markers in predicting the toxicity associated with novel targeted agents' administration is still controversial due to the heterogeneity of published data. Pharmacogenomics guidelines have been published by some international scientific consortia such as the Clinical Pharmacogenomics Implementation Consortium (CPIC) and the Dutch Pharmacogenetics Working Group (DPWG) strongly suggesting a pre-treatment dose adjustment of irinotecan based on UGT1A1*28 genotype and of fluoropyrimidines based on some DPYD genetic variants, to increase treatment safety. However, these recommendations are still poorly applied at the patient's bedside. Several ongoing projects in the U.S. and Europe are currently evaluating how pharmacogenomics can be implemented successfully in daily clinical practice. The majority of drug-related adverse events are still unexplained, and a great deal of ongoing research is aimed at improving knowledge of the role of pharmacogenomics in increasing treatment safety. In this review, the issue of pre-treatment identification of CRC patients at risk of toxicity via the analysis of patients' genetic profiles is addressed. Available pharmacogenomics guidelines with ongoing efforts to implement them in clinical practice and new exploratory markers for clinical validation are described.
Collapse
Affiliation(s)
- Erika Cecchin
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico - National Cancer Institute, 33081 Aviano, Italy
| | - Elena De Mattia
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico - National Cancer Institute, 33081 Aviano, Italy
| | - Fabrizio Ecca
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico - National Cancer Institute, 33081 Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico - National Cancer Institute, 33081 Aviano, Italy.
| |
Collapse
|
47
|
Palmirotta R, Carella C, Silvestris E, Cives M, Stucci SL, Tucci M, Lovero D, Silvestris F. SNPs in predicting clinical efficacy and toxicity of chemotherapy: walking through the quicksand. Oncotarget 2018; 9:25355-25382. [PMID: 29861877 PMCID: PMC5982750 DOI: 10.18632/oncotarget.25256] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 04/07/2018] [Indexed: 12/19/2022] Open
Abstract
In the "precision medicine" era, chemotherapy still remains the backbone for the treatment of many cancers, but no affordable predictors of response to the chemodrugs are available in clinical practice. Single nucleotide polymorphisms (SNPs) are gene sequence variations occurring in more than 1% of the full population, and account for approximately 80% of inter-individual genomic heterogeneity. A number of studies have investigated the predictive role of SNPs of genes enrolled in both pharmacodynamics and pharmacokinetics of chemotherapeutics, but the clinical implementation of related results has been modest so far. Among the examined germline polymorphic variants, several SNPs of dihydropyrimidine dehydrogenase (DPYD) and uridine diphosphate glucuronosyltransferases (UGT) have shown a robust role as predictors of toxicity following fluoropyrimidine- and/or irinotecan-based treatments respectively, and a few guidelines are mandatory in their detection before therapy initiation. Contrasting results, however, have been reported on the capability of variants of other genes as MTHFR, TYMS, ERCC1, XRCC1, GSTP1, CYP3A4/3A5 and ABCB1, in predicting either therapy efficacy or toxicity in patients undergoing treatment with pyrimidine antimetabolites, platinum derivatives, irinotecan and taxanes. While formal recommendations for routine testing of these SNPs cannot be drawn at this moment, therapeutic decisions may indeed benefit of germline genomic information, when available. Here, we summarize the clinical impact of germline genomic variants on the efficacy and toxicity of major chemodrugs, with the aim to facilitate the therapeutic expectance of clinicians in the odiern quicksand field of complex molecular biology concepts and controversial trial data interpretation.
Collapse
Affiliation(s)
- Raffaele Palmirotta
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Claudia Carella
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Erica Silvestris
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Mauro Cives
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Stefania Luigia Stucci
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Marco Tucci
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Domenica Lovero
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Franco Silvestris
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| |
Collapse
|
48
|
De Mattia E, Cecchin E, Montico M, Labriet A, Guillemette C, Dreussi E, Roncato R, Bignucolo A, Buonadonna A, D'Andrea M, Coppola L, Lonardi S, Lévesque E, Jonker D, Couture F, Toffoli G. Association of STAT-3 rs1053004 and VDR rs11574077 With FOLFIRI-Related Gastrointestinal Toxicity in Metastatic Colorectal Cancer Patients. Front Pharmacol 2018; 9:367. [PMID: 29706892 PMCID: PMC5908896 DOI: 10.3389/fphar.2018.00367] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 03/28/2018] [Indexed: 12/26/2022] Open
Abstract
Pharmacogenomics has largely been applied to the personalization of irinotecan-based treatment, focusing mainly on the study of genetic variants in adsorption, distribution, metabolism, and excretion (ADME) genes. The transcriptional control of ADME gene expression is mediated by a set of nuclear factors responding to cancer-related inflammation, which could have pharmacological implications. The aim of the present study was to uncover novel genetic predictors of neutropenia and gastrointestinal toxicity risk among 246 haplotype-tagging polymorphisms in 22 genes encoding inflammation-related cytokines and transcriptional regulators of ADME genes. The study comprised overall more than 400 metastatic colorectal cancer (mCRC) patients treated with first-line FOLFIRI, grouped in a discovery and a replication cohorts. A concordant protective effect of STAT-3 rs1053004 polymorphism against the risk of grade 3–4 gastrointestinal toxicity was observed in both the cohorts of patients (OR = 0.51, p = 0.045, q = 0.521 and OR = 0.39, p = 0.043, respectively). VDR rs11574077 polymorphism was demonstrated to affect both irinotecan biliary index (BI) and glucuronidation ratio (GR) by a pharmacokinetic analysis. This effect was consistent with an increased risk of grade 3–4 gastrointestinal toxicity in the discovery cohort (OR = 4.46, p = 0.010, q = 0.305). The association was not significant in the replication cohort (OR = 1.44, p = 0.601). These findings suggest an effect of STAT-3 and VDR polymorphisms on FOLFIRI-related gastrointestinal toxicity. If prospectively validated as predictive markers, they could be used to improve the clinical management of mCRC.
Collapse
Affiliation(s)
- Elena De Mattia
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | - Erika Cecchin
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | - Marcella Montico
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | - Adrien Labriet
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec, Research Center and Faculty of Pharmacy, Laval University, Québec, QC, Canada
| | - Chantal Guillemette
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec, Research Center and Faculty of Pharmacy, Laval University, Québec, QC, Canada
| | - Eva Dreussi
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | - Rossana Roncato
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | - Alessia Bignucolo
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | - Angela Buonadonna
- Medical Oncology Unit, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | - Mario D'Andrea
- Medical Oncology Unit, San Filippo Neri Hospital, Rome, Italy
| | - Luigi Coppola
- Pathology Unit, San Filippo Neri Hospital, Rome, Italy
| | - Sara Lonardi
- Medical Oncology Unit 1, Istituto Oncologico Veneto, Istituto Di Ricovero e Cura a Carattere Scientifico, Padua, Italy
| | - Eric Lévesque
- Centre Hospitalier Universitaire de Québec Research Center, Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Derek Jonker
- Division of Medical Oncology, Department of Medicine, Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| | - Félix Couture
- Centre Hospitalier Universitaire de Québec Research Center, Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Giuseppe Toffoli
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| |
Collapse
|
49
|
The reversal effect of Ginsenoside Rh2 on drug resistance in human colorectal carcinoma cells and its mechanism. Hum Cell 2018; 31:189-198. [DOI: 10.1007/s13577-017-0189-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 10/23/2017] [Indexed: 12/22/2022]
|
50
|
Lin Y, Chen Q, Liu QX, Zhou D, Lu X, Deng XF, Yang H, Zheng H, Qiu Y. High expression of DJ-1 promotes growth and invasion via the PTEN-AKT pathway and predicts a poor prognosis in colorectal cancer. Cancer Med 2018; 7:809-819. [PMID: 29441725 PMCID: PMC5852339 DOI: 10.1002/cam4.1325] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 12/14/2022] Open
Abstract
Cancer cell invasion and unlimited proliferation are key factors in patients with colorectal cancer (CRC). Increased protein deglycase DJ-1 in cancer cells is known to promote tumor growth; however, its role in CRC progression is not well defined. In this study, we investigated 100 CRC patients with disease stages I-IV to determine whether DJ-1 could serve as a prognostic biomarker in CRC. These results showed that DJ-1 expression in CRC tissues was higher than that in normal colon tissues and was associated with the (Tumor Node Metastasis) TNM stage. CRC patients with low DJ-1 expression had a longer overall survival than those with high expression, and multivariate and univariate analyses indicated that DJ-1 expression was an independent prognostic factor for overall survival in CRC. Furthermore, DJ-1 overexpression in two colon cancer cell lines, HCT116 and SW480, activated protein kinase AKT and downregulated tumor suppressor PTEN, whereas DJ-1 knockdown upregulated PTEN expression and effectively suppressed CRC cell invasion and proliferation both in vitro and in vivo, revealing a mechanism underlying DJ-1 pro-oncogenic activity in CRC. Treatment of MK2206, the specific AKT inhibitor, significantly decreased DJ-1-mediated cell proliferation and mobility in vitro. Taken together, these results suggest that DJ-1 may be a novel prognostic biomarker and potential therapeutic target in human CRC.
Collapse
Affiliation(s)
- Yong Lin
- Department of Pathology, The first affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Qian Chen
- Department of Pathology, The first affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Quan-Xing Liu
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Dong Zhou
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Xiao Lu
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Xu-Feng Deng
- Department of Cardiothoracic Surgery, First People's Hospital of Zunyi, Guizhou, 563000, China
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, The Third Military Medical University, Chongqing, 400037, China
| | - Hong Zheng
- Department of Thoracic Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Yuan Qiu
- Department of General Surgery, Xinqiao Hospital, The Third Military Medical University, Chongqing, 400037, China
| |
Collapse
|