1
|
Duan XP, Qin BD, Jiao XD, Liu K, Wang Z, Zang YS. New clinical trial design in precision medicine: discovery, development and direction. Signal Transduct Target Ther 2024; 9:57. [PMID: 38438349 PMCID: PMC10912713 DOI: 10.1038/s41392-024-01760-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 03/06/2024] Open
Abstract
In the era of precision medicine, it has been increasingly recognized that individuals with a certain disease are complex and different from each other. Due to the underestimation of the significant heterogeneity across participants in traditional "one-size-fits-all" trials, patient-centered trials that could provide optimal therapy customization to individuals with specific biomarkers were developed including the basket, umbrella, and platform trial designs under the master protocol framework. In recent years, the successive FDA approval of indications based on biomarker-guided master protocol designs has demonstrated that these new clinical trials are ushering in tremendous opportunities. Despite the rapid increase in the number of basket, umbrella, and platform trials, the current clinical and research understanding of these new trial designs, as compared with traditional trial designs, remains limited. The majority of the research focuses on methodologies, and there is a lack of in-depth insight concerning the underlying biological logic of these new clinical trial designs. Therefore, we provide this comprehensive review of the discovery and development of basket, umbrella, and platform trials and their underlying logic from the perspective of precision medicine. Meanwhile, we discuss future directions on the potential development of these new clinical design in view of the "Precision Pro", "Dynamic Precision", and "Intelligent Precision". This review would assist trial-related researchers to enhance the innovation and feasibility of clinical trial designs by expounding the underlying logic, which be essential to accelerate the progression of precision medicine.
Collapse
Affiliation(s)
- Xiao-Peng Duan
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Bao-Dong Qin
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xiao-Dong Jiao
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ke Liu
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhan Wang
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yuan-Sheng Zang
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
2
|
Tao J, Sun D, Zhou H, Zhu J, Zhang X, Hou H. Next-generation sequencing identifies potential novel therapeutic targets in Chinese HGSOC patients. Pathol Res Pract 2022; 238:154074. [PMID: 35988354 DOI: 10.1016/j.prp.2022.154074] [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] [Received: 02/27/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Targeted therapy, especially the use of poly (adenosine diphosphate ribose) polymerase (PARP) inhibitors (PARPis), has improved the outcome of patients with ovarian cancer. However, most high-grade serous ovarian cancer (HGSOC) patients have wild-type BRCA1/2, and it is necessary to disclose more potential novel targets for other available targeted drugs. So, detection of genetic alterations beyond BRCA1/2 is critical to screen HGSOC patients for personalized therapy. In this study, a broad, hybrid capture-based next-generation sequencing (NGS) assay was used to identify actionable genetic alterations from HGSOC cancer tissues. METHODS Sixty-eight patients with HGSOC were enrolled, including 6 International Federation of Gynecology and Obstetrics (FIGO) stage I, 15 stage II, 37 stage III and 10 stage IV patients. All patients signed informed consent forms. Potentially actionable genetic alterations, including base substitutions, indels, copy number alterations, and gene fusions, were identified using targeted NGS. RESULTS In our study, 14.7% (10/68) of the tumors harbored actionable genetic alterations in patients with BRCA1. A total of 25.0% (17/68) of patients without BRCA1 mutations harbored other actionable genetic alterations, such as homologous recombination repair (HRR) pathway-related genes (ATM, CDK12, FANCA, and FANCD2), PI3K/AKT/mTOR pathway genes (NF1, FBXW7, PIK3CA, PTEN, TSC1, and TSC2), and some other genes (ARID1A, FGFR1, KRAS, and NRAS). Furthermore, some patients harboring ARID1A or NF1 actionable genetic alterations showed good clinical efficacy to immune checkpoint inhibitors (ICIs) and everolimus, respectively. CONCLUSIONS Our research indicates that 39.7% (27/68) of patients with HGSOC harbored at least one actionable genetic alteration. 25.0% (17/68) of patients had somatic mutations or copy number variations beyond BRCA1 mutations and might be treated with off-label therapy or to be allocated into clinical trial. NGS assays of HGSOC patients are necessary to screen actionable genetic alterations to guide personalized and precise treatment.
Collapse
Affiliation(s)
- Junyan Tao
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266000, 59 Haier Road, Shandong 266000, China
| | - Dantong Sun
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266000, 59 Haier Road, Shandong 266000, China
| | - Hai Zhou
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266000, 59 Haier Road, Shandong 266000, China
| | - Jingjuan Zhu
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266000, 59 Haier Road, Shandong 266000, China
| | - Xiaochun Zhang
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266000, 59 Haier Road, Shandong 266000, China
| | - Helei Hou
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao 266000, 59 Haier Road, Shandong 266000, China.
| |
Collapse
|
3
|
Zhang Y, Liang S, Xiao B, Hu J, Pang Y, Liu Y, Yang J, Ao J, Wei L, Luo X. MiR-323a regulates ErbB3/EGFR and blocks gefitinib resistance acquisition in colorectal cancer. Cell Death Dis 2022; 13:256. [PMID: 35319011 PMCID: PMC8940899 DOI: 10.1038/s41419-022-04709-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 02/15/2022] [Accepted: 03/03/2022] [Indexed: 12/12/2022]
Abstract
The rapid onset of resistance to epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) limits its clinical utility in colorectal cancer (CRC) patients, and pan-erb-b2 receptor tyrosine kinase (ErbB) treatment strategy may be the alternative solution. The aim of this study was to develop a possible microRNA multi-ErbB treatment strategy to overcome EGFR-TKI resistance. We detect the receptor tyrosine kinase activity in gefitinib-resistant colorectal cancer cells, ErbB3/EGFR is significantly activated and provides a potential multi-ErbB treatment target. MiR-323a-3p, a tumor suppressor, could target both ErbB3 and EGFR directly. Apoptosis is the miR-323a-3p inducing main biological process by functional enrichment analysis, and The EGFR and ErbB signaling are the miR-323a-3p inducing main pathway by KEGG analysis. MiR-323a-3p promotes CRC cells apoptosis by targeting ErbB3-phosphoinositide 3-kinases (PI3K)/PKB protein kinase (Akt)/glycogen synthase kinase 3 beta (GSK3β)/EGFR-extracellular regulated MAP kinase (Erk1/2) signaling directly. And miR-323a-3p, as a multi-ErbBs inhibitor, increase gefitinib sensitivity of the primary cell culture from combination miR-323a-3p and gefitinib treated subcutaneous tumors. MiR-323a-3p reverses ErbB3/EGFR signaling activation in gefitinib-resistant CRC cell lines and blocks acquired gefitinib resistance.
Collapse
Affiliation(s)
- Yuanzhou Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, People's Republic of China
| | - Shunshun Liang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, People's Republic of China
| | - Bowen Xiao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, People's Republic of China
| | - Jingying Hu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, People's Republic of China
| | - Yechun Pang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, People's Republic of China
| | - Yuling Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, People's Republic of China
| | - Juan Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, People's Republic of China
| | - Junpin Ao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, People's Republic of China
| | - Lin Wei
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, People's Republic of China
| | - Xiaoying Luo
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
4
|
Yeoh Y, Low TY, Abu N, Lee PY. Regulation of signal transduction pathways in colorectal cancer: implications for therapeutic resistance. PeerJ 2021; 9:e12338. [PMID: 34733591 PMCID: PMC8544255 DOI: 10.7717/peerj.12338] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022] Open
Abstract
Resistance to anti-cancer treatments is a critical and widespread health issue that has brought serious impacts on lives, the economy and public policies. Mounting research has suggested that a selected spectrum of patients with advanced colorectal cancer (CRC) tend to respond poorly to both chemotherapeutic and targeted therapeutic regimens. Drug resistance in tumours can occur in an intrinsic or acquired manner, rendering cancer cells insensitive to the treatment of anti-cancer therapies. Multiple factors have been associated with drug resistance. The most well-established factors are the emergence of cancer stem cell-like properties and overexpression of ABC transporters that mediate drug efflux. Besides, there is emerging evidence that signalling pathways that modulate cell survival and drug metabolism play major roles in the maintenance of multidrug resistance in CRC. This article reviews drug resistance in CRC as a result of alterations in the MAPK, PI3K/PKB, Wnt/β-catenin and Notch pathways.
Collapse
Affiliation(s)
- Yeelon Yeoh
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Teck Yew Low
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nadiah Abu
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Pey Yee Lee
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
5
|
A human antibody against human endothelin receptor type A that exhibits antitumor potency. Exp Mol Med 2021; 53:1437-1448. [PMID: 34588605 PMCID: PMC8492878 DOI: 10.1038/s12276-021-00678-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/11/2021] [Accepted: 07/19/2021] [Indexed: 12/22/2022] Open
Abstract
Endothelin receptor A (ETA), a class A G-protein-coupled receptor (GPCR), is involved in the progression and metastasis of colorectal, breast, lung, ovarian, and prostate cancer. We overexpressed and purified human endothelin receptor type A in Escherichia coli and reconstituted it with lipid and membrane scaffold proteins to prepare an ETA nanodisc as a functional antigen with a structure similar to that of native GPCR. By screening a human naive immune single-chain variable fragment phage library constructed in-house, we successfully isolated a human anti-ETA antibody (AG8) exhibiting high specificity for ETA in the β-arrestin Tango assay and effective inhibitory activity against the ET-1-induced signaling cascade via ETA using either a CHO-K1 cell line stably expressing human ETA or HT-29 colorectal cancer cells, in which AG8 exhibited IC50 values of 56 and 51 nM, respectively. In addition, AG8 treatment repressed the transcription of inhibin βA and reduced the ETA-induced phosphorylation of protein kinase B and extracellular regulated kinase. Furthermore, tumor growth was effectively inhibited by AG8 in a colorectal cancer mouse xenograft model. The human anti-ETA antibody isolated in this study could be used as a potential therapeutic for cancers, including colorectal cancer. A therapeutic antibody that targets a receptor involved in cancer progression shows significant anti-cancer effects in trials in mice. Endothelin receptor A (ETA) promotes the progression and metastasis of several cancers, and patients with high ETA expression often have poor survival rates. Several small molecule drugs that target ETA are currently undergoing trials. Now, Sang Taek Jung at the Korea University in Seoul, together with scientists across South Korea, have identified and isolated a human antibody that specifically binds to ETA. The team developed an antigen that mimics ETA, and identified and isolated the antibody it bound to. The antibody exhibited potent anti-tumor effects in cell cultures and trials in mice. Such therapeutic antibodies show higher affinity for their targets than other drugs, resulting in fewer side effects and higher efficacy.
Collapse
|
6
|
Fang K, Zhan Y, Zhu R, Wang Y, Wu C, Sun M, Qiu Y, Yuan Z, Liang X, Yin P, Xu K. Bufalin suppresses tumour microenvironment-mediated angiogenesis by inhibiting the STAT3 signalling pathway. J Transl Med 2021; 19:383. [PMID: 34496870 PMCID: PMC8424978 DOI: 10.1186/s12967-021-03058-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/26/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Antiangiogenic therapy has increasingly become an important strategy for the treatment of colorectal cancer. Recent studies have shown that the tumour microenvironment (TME) promotes tumour angiogenesis. Bufalin is an active antitumour compound whose efficacy has been indicated by previous studies. However, there are very few studies on the antiangiogenic effects of bufalin. METHODS Herein, human umbilical vein endothelial cell (HUVEC) tube formation, migration and adhesion tests were used to assess angiogenesis in vitro. Western blotting and quantitative PCR were used to detect relevant protein levels and mRNA expression levels. A subcutaneous xenograft tumour model and a hepatic metastasis model were established in mice to investigate the influence of bufalin on angiogenesis mediated by the TME in vivo. RESULTS We found that angiogenesis mediated by cells in the TME was significantly inhibited in the presence of bufalin. The results demonstrated that the proangiogenic genes in HUVECs, such as VEGF, PDGFA, E-selectin and P-selectin, were downregulated by bufalin and that this downregulation was mediated by inhibition of the STAT3 pathway. Overexpression of STAT3 reversed the inhibitory effects of bufalin on angiogenesis. Furthermore, there was little reduction in angiogenesis when bufalin directly acted on the cells in the tumour microenvironment. CONCLUSION Our findings demonstrate that bufalin suppresses tumour microenvironment-mediated angiogenesis by inhibiting the STAT3 signalling pathway in vascular endothelial cells, revealing that bufalin may be used as a new antiangiogenic adjuvant therapy medicine to treat colorectal cancer.
Collapse
Affiliation(s)
- Kai Fang
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444 China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Rd, Shanghai, 200237 China
| | - Yueping Zhan
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
| | - Ruiqiu Zhu
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, 230032 China
| | - Yuqian Wang
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
| | - Chengqi Wu
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444 China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Rd, Shanghai, 200237 China
| | - Min Sun
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
| | - Yanyan Qiu
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
| | - Zeting Yuan
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, 230032 China
| | - Xin Liang
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Rd, Shanghai, 200237 China
| | - Peihao Yin
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, 230032 China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444 China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062 China
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, 230032 China
- Wenzhou Institute of Shanghai University, Wenzhou, 325000 China
| |
Collapse
|
7
|
Cetuximab-Mediated Protection from Hypoxia- Induced Cell Death: Implications for Therapy Sequence in Colorectal Cancer. Cancers (Basel) 2020; 12:cancers12103050. [PMID: 33092032 PMCID: PMC7589936 DOI: 10.3390/cancers12103050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Therapeutic antibodies are an integral part of treatment regimens for metastasized colorectal cancer. In KRAS wildtype tumors both bevacizumab and cetuximab are active. While bevacizumab has previously been shown to induce tumor hypoxia, we here report that EGFR inhibition by cetuximab protects colon cancer cells from hypoxia-induced cell death. This effect appears to be responsible for the inferior efficacy of a treatment sequence of bevacizumab followed by cetuximab versus an inverse sequence that we observed in a colorectal cancer mouse model. It also offers a mechanistic explanation for effects observed in clinical trials such as underadditive or even detrimental effects when combining bevacizumab and cetuximab (CAIRO2 trial) and the superior efficacy of first line cetuximab (FIRE-3 trial) under chemotherapy backbones in colorectal cancer. Abstract Monoclonal antibodies like cetuximab, targeting the epidermal growth factor receptor (EGFR), and bevacizumab, targeting the vascular endothelial growth factor (VEGF), are an integral part of treatment regimens for metastasized colorectal cancer. However, inhibition of the EGFR has been shown to protect human glioma cells from cell death under hypoxic conditions. In colon carcinoma cells, the consequences of EGFR blockade in hypoxia (e.g., induced by bevacizumab) have not been evaluated yet. LIM1215 and SW948 colon carcinoma and LNT-229 glioblastoma cells were treated with cetuximab, PD153035, and erlotinib and analyzed for cell density and viability. The sequential administration of either cetuximab followed by bevacizumab (CET->BEV) or bevacizumab followed by cetuximab (BEV->CET) was investigated in a LIM1215 (KRAS wildtype) and SW948 (KRAS mutant) xenograft mouse model. In vitro, cetuximab protected from hypoxia. In the LIM1215 model, a survival benefit with cetuximab and bevacizumab monotherapy was observed, but only the sequence CET->BEV showed an additional benefit. This effect was confirmed in the SW948 model. Our observations support the hypothesis that bevacizumab modulates the tumor microenvironment (e.g., by inducing hypoxia) where cetuximab could trigger protective effects when administered later on. The sequence CET->BEV therefore seems to be superior as possible mutual adverse effects are bypassed.
Collapse
|
8
|
Huang Z, Liu J, Luo L, Sheng P, Wang B, Zhang J, Peng SS. Genome-Wide Identification of a Novel Autophagy-Related Signature for Colorectal Cancer. Dose Response 2019; 17:1559325819894179. [PMID: 31853237 PMCID: PMC6906358 DOI: 10.1177/1559325819894179] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 11/12/2019] [Accepted: 11/19/2019] [Indexed: 02/06/2023] Open
Abstract
Background: Plenty of evidence has suggested that autophagy plays a crucial role in the
biological processes of cancers. This study aimed to screen
autophagy-related genes (ARGs) and establish a novel a scoring system for
colorectal cancer (CRC). Methods: Autophagy-related genes sequencing data and the corresponding clinical data
of CRC in The Cancer Genome Atlas were used as training data set. The
GSE39582 data set from the Gene Expression Omnibus was used as validation
set. An autophagy-related signature was developed in training set using
univariate Cox analysis followed by stepwise multivariate Cox analysis and
assessed in the validation set. Then we analyzed the function and pathways
of ARGs using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes
(KEGG) database. Finally, a prognostic nomogram combining the
autophagy-related risk score and clinicopathological characteristics was
developed according to multivariate Cox analysis. Results: After univariate and multivariate analysis, 3 ARGs were used to construct
autophagy-related signature. The KEGG pathway analyses showed several
significantly enriched oncological signatures, such as p53 signaling
pathway, apoptosis, human cytomegalovirus infection, platinum drug
resistance, necroptosis, and ErbB signaling pathway. Patients were divided
into high- and low-risk groups, and patients with high risk had
significantly shorter overall survival (OS) than low-risk patients in both
training set and validation set. Furthermore, the nomogram for predicting 3-
and 5-year OS was established based on autophagy-based risk score and
clinicopathologic factors. The area under the curve and calibration curves
indicated that the nomogram showed well accuracy of prediction. Conclusions: Our proposed autophagy-based signature has important prognostic value and may
provide a promising tool for the development of personalized therapy.
Collapse
Affiliation(s)
- Zhi Huang
- Department of General Surgery, Dazhou Central Hospital, Sichuan, People's Republic of China
| | - Jie Liu
- Department of General Surgery, Dazhou Central Hospital, Sichuan, People's Republic of China
| | - Liang Luo
- Department of General Surgery, Dazhou Central Hospital, Sichuan, People's Republic of China
| | - Pan Sheng
- Department of General Surgery, Dazhou Central Hospital, Sichuan, People's Republic of China
| | - Biao Wang
- Department of General Surgery, Dazhou Central Hospital, Sichuan, People's Republic of China
| | - Jun Zhang
- Department of General Surgery, Dazhou Central Hospital, Sichuan, People's Republic of China
| | - Sha-Sha Peng
- Department of Hepatobiliary and Pancreatic Surgery, Huangshi Central Hospital of Edong Healthcare Group, Hubei Polytechnic University, Hubei, People's Republic of China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
9
|
Cardone C, Martinelli E, Troiani T, Sforza V, Avallone A, Nappi A, Montesarchio V, Andreozzi F, Biglietto M, Calabrese F, Bordonaro R, Cordio S, Bregni G, Febbraro A, Garcia-Carbonero R, Feliu J, Cervantes A, Ciardiello F. Exploratory findings from a prematurely closed international, multicentre, academic trial: RAVELLO, a phase III study of regorafenib versus placebo as maintenance therapy after first-line treatment in RAS wild-type metastatic colorectal cancer. ESMO Open 2019; 4:e000519. [PMID: 31555481 PMCID: PMC6735666 DOI: 10.1136/esmoopen-2019-000519] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/20/2019] [Accepted: 05/22/2019] [Indexed: 01/07/2023] Open
Abstract
Background In patients with RAS wild-type (WT) metastatic colorectal cancer (mCRC), the role of maintenance therapy after first-line treatment with chemotherapy plus antiepidermal growth factor receptor (EGFR) monoclonal antibodies (MoAb) is still an object of debate. Methods We assessed the efficacy and safety of regorafenib as a switch maintenance strategy after upfront 5-fluorouracil-based chemotherapy plus an anti-EGFR MoAb in patients with RAS WT mCRC. RAVELLO was a phase III, international, double-blind, placebo-controlled, academic trial. The primary endpoint was progression-free survival (PFS). Secondary endpoints included overall survival and toxicity. Regorafenib or placebo were administered daily for 3 weeks of 4-week cycle until disease progression or unacceptable toxicity, up to 24 months. Results The study was stopped prematurely due to slow accrual and lack of funding after the randomisation of 21 patients: 11 in the regorafenib arm and 10 in the placebo arm. The small sample size precludes any statistical analysis. Toxicity was acceptable and consistent with the known regorafenib safety profile. Median PFS was similar in the two arms. However, a subgroup of patients treated with regorafenib experienced a remarkably long PFS. Three patients were progression free at 9 months in the regorafenib arm versus one patient in the placebo arm, whereas at 12 months two regorafenib-treated patients were still progression free versus none in the placebo arm. Conclusion RAVELLO trial demonstrated that growing financial and bureaucratic hurdles affect the feasibility of independent academic research. Although stopped prematurely and within the limited sample size, RAVELLO suggests that regorafenib has not a major activity in maintenance setting after upfront chemotherapy and anti-EGFR MoAb. However, a subgroup of patients experienced a remarkable long PFS, indicating that a better refinement of the patient population would help to identify subjects that might benefit from a regorafenib personalised approach in the switch maintenance setting.
Collapse
Affiliation(s)
- Claudia Cardone
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Erika Martinelli
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Teresa Troiani
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Vincenzo Sforza
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| | - Antonio Avallone
- Department of Experimental Medicine, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Anna Nappi
- Department of Experimental Medicine, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | | | - Francesca Andreozzi
- Azienda Ospedaliera dei Colli, Medical Oncology, Monaldi Hospital, Napoli, Italy
| | - Maria Biglietto
- Medical Oncology, Antonio Cardarelli Hospital, Napoli, Italy
| | | | - Roberto Bordonaro
- Medical Oncology, National Specialist Hospital Garibaldi, Catania, Italy
| | - Stefano Cordio
- Medical Oncology, National Specialist Hospital Garibaldi, Catania, Italy
| | | | - Antonio Febbraro
- Medical Oncology, Hospital Sacro Cuore di Gesù, Fatebenefratelli, Benevento, Italy
| | - Rocio Garcia-Carbonero
- Medical Oncology, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), UCM, CNIO, CIBERONC, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Jaime Feliu
- CIBERONC, Medical Oncology, Hospital Universitario La Paz, Madrid, Spain
| | - Andrés Cervantes
- CIBERONC, Medical Oncology, Biomedical Research Institute INCLIVA, University of Valencia, Valencia, Spain
| | - Fortunato Ciardiello
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Napoli, Campania, Italy
| |
Collapse
|
10
|
Zoetemelk M, Rausch M, Colin DJ, Dormond O, Nowak-Sliwinska P. Short-term 3D culture systems of various complexity for treatment optimization of colorectal carcinoma. Sci Rep 2019; 9:7103. [PMID: 31068603 PMCID: PMC6506470 DOI: 10.1038/s41598-019-42836-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 04/10/2019] [Indexed: 02/07/2023] Open
Abstract
Three-dimensional (3D) cultures have the potential to increase the predictive value of pre-clinical drug research and bridge the gap towards anticipating clinical outcome of proposed treatments. However, their implementation in more advanced drug-discovery programs is still in its infancy due to the lack of reproducibility and low time- and cost effectiveness. HCT116, SW620 and DLD1 cells, cell lines with distinct mutations, grade and origin, were co-cultured with fibroblasts and endothelial cells (EC) in 3D spheroids. Clinically relevant drugs, i.e. 5-fluorouracil (5−FU), regorafenib and erlotinib, were administered individually to in CRC cell cultures. In this study, we established a robust, low-cost and reproducible short-term 3D culture system addressing the various complexities of the colorectal carcinoma (CRC) microenvironment. We observed a dose-dependent increase of erlotinib sensitivity in 3D (co-)cultures compared to 2D cultures. Furthermore, we compared the drug combination efficacy and drug-drug interactions administered in 2D, 3D and 3D co-cultures. We observed that synergistic/additive drug-drug interactions for drug combinations administered at low doses shifted towards additive and antagonistic when applied at higher doses in metastatic CRC cells. The addition of fibroblasts at various ratios and EC increased the resistance to some drug combinations in SW620 and DLD1 cells, but not in HCT116. Retreatment of SW620 3D co-cultures with a low-dose 3-drug combination was as active (88% inhibition, relative to control) as 5-FU treatment at high dose (100 μM). Moreover, 3D and 3D co-cultures responded variably to the drug combination treatments, and also signalling pathways were differently regulated, probably due to the influence of fibroblasts and ECs on cancer cells. The short-term 3D co-culture system developed here is a powerful platform for screening (combination) therapies. Understanding of signalling in 3D co-cultures versus 3D cultures and the responses in the 3D models upon drug treatment might be beneficial for designing anti-cancer therapies.
Collapse
Affiliation(s)
- Marloes Zoetemelk
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 1211, Geneva 4, Switzerland.,Translational Research Center in Oncohaematology, 1211, Geneva 4, Switzerland
| | - Magdalena Rausch
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 1211, Geneva 4, Switzerland.,Translational Research Center in Oncohaematology, 1211, Geneva 4, Switzerland
| | - Didier J Colin
- Centre for BioMedical Imaging (CIBM), University Hospitals and University of Geneva, 1211, Geneva 4, Switzerland
| | - Olivier Dormond
- Department of Visceral Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Patrycja Nowak-Sliwinska
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 1211, Geneva 4, Switzerland. .,Translational Research Center in Oncohaematology, 1211, Geneva 4, Switzerland.
| |
Collapse
|
11
|
Inhibition of SIRT2 limits tumour angiogenesis via inactivation of the STAT3/VEGFA signalling pathway. Cell Death Dis 2018; 10:9. [PMID: 30584257 PMCID: PMC6315023 DOI: 10.1038/s41419-018-1260-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/24/2018] [Accepted: 11/30/2018] [Indexed: 12/28/2022]
Abstract
Mounting evidence has demonstrated that angiogenesis plays an important role in tumour progression. However, the key regulators in tumour angiogenesis remain unclear. Recently, emerging reports have indicated that SIRT2 plays critical roles in proliferation, metastasis and tumourigenesis in diverse tumours. However, the function of SIRT2 in tumour angiogenesis and the mechanism underlying the regulation of angiogenesis by SIRT2 are still unknown. Here, we found that SIRT2 was upregulated in colorectal cancer tissues compared to that in normal samples and that the elevated SIRT2 was associated with poor prognosis in patients with colorectal cancer. In addition, a series of in vitro and in vivo experiments were performed to demonstrate the role of SIRT2 in tumour angiogenesis. We showed that silencing SIRT2 significantly suppressed tumour angiogenesis. Mechanistically, the knockdown of SIRT2 inhibited STAT3 phosphorylation, causing decreased secretion of VEGFA. Notably, we found that SIRT2 directly interacted with STAT3 and affected the phosphorylation of STAT3 and the translocation of phosphorylated STAT3 to the nucleus. Importantly, a series of rescue experiments suggested that the function of SIRT2 in tumour angiogenesis depends on the STAT3/VEGFA signalling pathway. Our findings provide insight into the important role of SIRT2 in colon tumour angiogenesis and suggest that SIRT2/STAT3/VEGFA might be a novel prognostic biomarker and a potential therapeutic target for patients with colorectal cancer.
Collapse
|