1
|
Sobolewski C, Legrand N. Celecoxib Analogues for Cancer Treatment: An Update on OSU-03012 and 2,5-Dimethyl-Celecoxib. Biomolecules 2021; 11:biom11071049. [PMID: 34356673 PMCID: PMC8302000 DOI: 10.3390/biom11071049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 12/20/2022] Open
Abstract
Cyclooxygenase-2 (COX-2) is an important enzyme involved in prostaglandins biosynthesis from arachidonic acid. COX-2 is frequently overexpressed in human cancers and plays a major tumor promoting function. Accordingly, many efforts have been devoted to efficiently target the catalytic site of this enzyme in cancer cells, by using COX-2 specific inhibitors such as celecoxib. However, despite their potent anti-tumor properties, the myriad of detrimental effects associated to the chronic inhibition of COX-2 in healthy tissues, has considerably limited their use in clinic. In addition, increasing evidence indicate that these anti-cancerous properties are not strictly dependent on the inhibition of the catalytic site. These findings have led to the development of non-active COX-2 inhibitors analogues aiming at preserving the antitumor effects of COX-2 inhibitors without their side effects. Among them, two celecoxib derivatives, 2,5-Dimethyl-Celecoxib and OSU-03012, have been developed and suggested for the treatment of viral (e.g., recently SARS-CoV-2), inflammatory, metabolic diseases and cancers. These molecules display stronger anti-tumor properties than celecoxib and thus may represent promising anti-cancer molecules. In this review, we discuss the impact of these two analogues on cancerous processes but also their potential for cancer treatment alone or in combination with existing approaches.
Collapse
Affiliation(s)
- Cyril Sobolewski
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
- Correspondence: ; Tel.: +41-22-379-5421
| | - Noémie Legrand
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland;
| |
Collapse
|
2
|
Non-Steroidal Anti-Inflammatory Drugs Increase Cisplatin, Paclitaxel, and Doxorubicin Efficacy against Human Cervix Cancer Cells. Pharmaceuticals (Basel) 2020; 13:ph13120463. [PMID: 33333716 PMCID: PMC7765098 DOI: 10.3390/ph13120463] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 12/26/2022] Open
Abstract
This study shows that the non-steroidal anti-inflammatory drug (NSAID) celecoxib and its non-cyclooxygenase-2 (COX2) analogue dimethylcelecoxib (DMC) exert a potent inhibitory effect on the growth of human cervix HeLa multi-cellular tumor spheroids (MCTS) when added either at the beginning (“preventive protocol”; IC50 = 1 ± 0.3 nM for celecoxib and 10 ± 2 nM for DMC) or after spheroid formation (“curative protocol”; IC50 = 7.5 ± 2 µM for celecoxib and 32 ± 10 µM for DMC). These NSAID IC50 values were significantly lower than those attained in bidimensional HeLa cells (IC50 = 55 ± 9 µM celecoxib and 48 ± 2 µM DMC) and bidimensional non-cancer cell cultures (3T3 fibroblasts and MCF-10A mammary gland cells with IC50 from 69 to >100 µM, after 24 h). The copper-based drug casiopeina II-gly showed similar potency against HeLa MCTS. Synergism analysis showed that celecoxib, DMC, and casiopeinaII-gly at sub-IC50 doses increased the potency of cisplatin, paclitaxel, and doxorubicin to hinder HeLa cell proliferation through a significant abolishment of oxidative phosphorylation in bidimensional cultures, with no apparent effect on non-cancer cells (therapeutic index >3.6). Similar results were attained with bidimensional human cervix cancer SiHa and human glioblastoma U373 cell cultures. In HeLa MCTS, celecoxib, DMC and casiopeina II-gly increased cisplatin toxicity by 41–85%. These observations indicated that celecoxib and DMC used as adjuvant therapy in combination with canonical anti-cancer drugs may provide more effective alternatives for cancer treatment.
Collapse
|
3
|
Chen Z, Chen Y, Peng L, Wang X, Tang N. 2,5-dimethylcelecoxib improves immune microenvironment of hepatocellular carcinoma by promoting ubiquitination of HBx-induced PD-L1. J Immunother Cancer 2020; 8:jitc-2020-001377. [PMID: 33028694 PMCID: PMC7542662 DOI: 10.1136/jitc-2020-001377] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2020] [Indexed: 12/17/2022] Open
Abstract
Background 2,5-dimethylcelecoxib (DMC) is a targeted inhibitor of microsomal prostaglandin E synthase-1 (mPGES-1), a key enzyme in the PGE2 synthesis pathway of inflammatory mediators. Previous studies have confirmed that DMC can inhibit the growth of hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC). However, it is not known whether DMC is involved in the changes of tumor immune microenvironment. Methods In this study, we explored the effects of DMC on HBV-related HCC immune microenvironment, and deeply analyzed its unique effect and mechanism on programmed death receptor 1 (PD-1)/and its ligand 1 (PD-L1) pathway. Results Clinical hepatoma tissues detection showed that compared with non-virus-related HCC, the level of CD8 of HBV-related HCC was significantly lower, while the levels of PD-L1 and CD163 were higher. In vivo experiments indicated that DMC could increase the level of tumor infiltrating CD8+ T cells in hepatitis B virus X (HBx) (+) hepatoma cells implanted mouse models, and inhibit the expression of PD-L1 and CD163 in tumor tissues. DMC combined with atezolizumab had more significant antitumor effect and stronger blocking effect on PD-1/PD-L1 pathway. Mechanism studies have shown that DMC can promote ubiquitin degradation of HBx-induced PD-L1 protein in HCC cells by activating adenosine 5′-monophosphate-activated protein kinase pathway. Further experiments confirmed that this process was mainly mediated by E3 ligase RBX1. Conclusions Our results uncover a role for DMC in promoting HBV-related HCC immune microenvironment, which not only enrich the relationship between inflammatory factors (mPGES-1/PGE2 pathway) and immunosuppression (PD-L1), but also provide an important strategic reference for multitarget or combined immunotherapy of HBV-related HCC.
Collapse
Affiliation(s)
- Zhanfei Chen
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yiyin Chen
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Lirong Peng
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaoqian Wang
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Nanhong Tang
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China .,Fujian Medical University Cancer Center, Fujian Medical University, Fuzhou, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| |
Collapse
|
4
|
Li S, Jiang M, Wang L, Yu S. Combined chemotherapy with cyclooxygenase-2 (COX-2) inhibitors in treating human cancers: Recent advancement. Biomed Pharmacother 2020; 129:110389. [PMID: 32540642 DOI: 10.1016/j.biopha.2020.110389] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/28/2020] [Accepted: 06/07/2020] [Indexed: 12/20/2022] Open
Abstract
Chemotherapy with a single chemotherapeutic agent or a combined chemotherapeutic regimen is the clinically standardized treatment for almost all human cancers. Upregulated expression of cyclooxygenase (COX)-2, also known as prostaglandin-endoperoxide synthase (PTGS), is associated with human carcinogenesis and cancer progression and COX-2 inhibitors show antitumor activity in different human cancers. Thus, a combination of chemotherapeutic agents with COX-2 inhibitors has been shown to improve therapeutic effects on human cancers. This review discusses and summarizes recent advances in cancer control and treatment using various antineoplastic drugs combined with COX-2 inhibitors. These combinations showed synergistic antitumor effects. At the gene level, COX-2 inhibitors can reduce inflammatory factors thereby regulating macrophage recruitment for activating the antitumor immune microenvironment; downregulating vascular endothelial growth factor (VEGF) to inhibit tumor angiogenesis; and inhibiting the PI3K/Akt signaling pathway to induce tumor cell apoptosis. In addition, such a combination can reduce toxicity and chemoresistance and enhance radiosensitivity, although COX-2 inhibitors-related cardiotoxicity may potentially affect its use. Further in-depth investigation of these drug combinations is needed to maximize antitumor efficacy and minimize the side effects.
Collapse
Affiliation(s)
- Shuangshuang Li
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Min Jiang
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Lu Wang
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacy, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250013, China.
| | - Shuwen Yu
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacy, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250013, China.
| |
Collapse
|
5
|
Won YS, Seo KI. Sanggenol L promotes apoptotic cell death in melanoma skin cancer cells through activation of caspase cascades and apoptosis-inducing factor. Food Chem Toxicol 2020; 138:111221. [PMID: 32084496 DOI: 10.1016/j.fct.2020.111221] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/22/2020] [Accepted: 02/17/2020] [Indexed: 12/18/2022]
Abstract
Sanggenol L is one component of root bark of Morus alba. The molecular and cellular mechanisms of sanggenol L effects on melanoma cells are not well known. Recently, melanoma is the most common skin cancer with a high mortality rate not only in United States, but also in East Asia. Therefore, safe and effective treatments for melanoma treatment are required. In this study, we investigated whether or not sanggenol L possesses anti-cancer activity in human and mouse melanoma skin cancer cells. Sanggenol L treatment exerted significant cell growth inhibitory effects and inhibited colony formation capacity against B16, SK-MEL-2, and SK-MEL-28 melanoma skin cancer cells, whereas HaCaT human epithelial keratinocyte cells was unaffected by sanggenol L treatment. Sanggenol L treatment resulted in apoptotic cell death in melanoma skin cancer cells, which was characterized by accumulation of apoptotic cells, nuclear condensation, and apoptotic bodies. We also showed that sanggenol L treatment induced caspase-dependent apoptosis (up-regulation of Bax and cleaved-PARP or down-regulation of Bid, Bcl-2, procaspse-3, -8, and -9), induction of caspase-independent apoptosis (up-regulation of AIF and Endo G on cytosol) in melanoma skin cancer cells. These results suggest that sanggenol L induces caspase-dependent and -independent apoptosis in melanoma skin cancer cells.
Collapse
Affiliation(s)
- Yeong-Seon Won
- Department of Biotechnology, Dong-A University, Busan, 49315, Republic of Korea
| | - Kwon-Il Seo
- Department of Biotechnology, Dong-A University, Busan, 49315, Republic of Korea.
| |
Collapse
|
6
|
Won YS, Seo KI. Lupiwighteone induces caspase-dependent and -independent apoptosis on human breast cancer cells via inhibiting PI3K/Akt/mTOR pathway. Food Chem Toxicol 2019; 135:110863. [PMID: 31604113 DOI: 10.1016/j.fct.2019.110863] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/04/2019] [Accepted: 09/27/2019] [Indexed: 12/25/2022]
Abstract
Breast cancer is one of the most common causes of mortality in women. Lupiwighteone has anticancer effects in prostate cancer cells and neuroblastoma cells. However, the molecular and cellular mechanisms of lupiwighteone effects on human breast cancer cells are not as well known. In the present study, we investigated the effects of lupiwighteone on the proliferation and apoptosis of two different human cancer cells; MCF-7, an estrogen receptor (ER)-positive human breast cancer cell, and MDA-MB-231, a triple negative human breast cancer cell. Lupiwighteone treatment decreased the viability of MCF-7 and MDA-MB-231 cells. Lupiwighteone treatment resulted in apoptotic cell death in breast cancer cells, which was characterized by DNA fragmentation, accumulation of apoptotic cells, and nuclear condensation. We also showed that treatment with lupiwighteone induced caspase-dependent apoptosis (up-regulation of caspase-3, -7, -8, -9, PARP, and Bax or down-regulation of Bid, Bcl-2), induction of caspase-independent apoptosis (up-regulation of AIF and Endo G on cytosol), and inhibition of the PI3K/Akt/mTOR signaling pathway (down-regulation of PI3K, p-Akt, and p-mTOR) in both MCF-7 and MDA-MB-231 cells. These results suggest that lupiwighteone induces caspase-dependent and -independent apoptosis in both breast cancer cell lines via inhibiting PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Yeong-Seon Won
- Department of Biotechnology, Dong-A University, Busan, 49315, Republic of Korea
| | - Kwon-Il Seo
- Department of Biotechnology, Dong-A University, Busan, 49315, Republic of Korea.
| |
Collapse
|
7
|
Zhang B, Wang L, Zhang Q, Yan Y, Jiang H, Hu R, Zhou X, Liu X, Feng J, Lin N. The Ibr-7 derivative of ibrutinib exhibits enhanced cytotoxicity against non-small cell lung cancer cells via targeting of mTORC1/S6 signaling. Mol Oncol 2019; 13:946-958. [PMID: 30663221 PMCID: PMC6441926 DOI: 10.1002/1878-0261.12454] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 12/23/2018] [Accepted: 01/05/2019] [Indexed: 12/16/2022] Open
Abstract
Ibrutinib is a small molecule drug that targets Bruton's tyrosine kinase in B-cell malignancies and is highly efficient at killing mantle cell lymphoma and chronic lymphocytic leukemia. However, the anti-cancer activity of ibrutinib against solid tumors, such as non-small cell lung cancer (NSCLC), remains low. To improve the cytotoxicity of ibrutinib towards lung cancer, we synthesized a series of ibrutinib derivatives, of which Ibr-7 exhibited superior anti-cancer activity to ibrutinib, especially against epithelial growth factor receptor (EGFR) wild-type NSCLC cell lines. Ibr-7 was observed to dramatically suppress the mammalian target of Rapamycin complex 1 (mTORC1)/S6 signaling pathway, which is only slightly affected by ibrutinib, thus accounting for the superior anti-cancer activity of Ibr-7 towards NSCLC. Ibr-7 was shown to overcome the elevation of Mcl-1 caused by ABT-199 mono-treatment, and thus exhibited a significant synergistic effect when combined with ABT-199. In conclusion, we used a molecular substitution method to generate a novel ibrutinib derivative, termed Ibr-7, which exhibits enhanced anti-cancer activity against NSCLC cells as compared with the parental compound.
Collapse
Affiliation(s)
- Bo Zhang
- Translational Medicine Research CenterAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouChina
- Affiliated Hangzhou First People's HospitalZhejiang Chinese Medical UniversityHangzhouChina
| | - Linling Wang
- Translational Medicine Research CenterAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouChina
- Affiliated Hangzhou First People's HospitalZhejiang Chinese Medical UniversityHangzhouChina
- Shaoxing Hospital of Traditional Chinese MedicineShaoxingChina
| | - Qi Zhang
- Translational Medicine Research CenterAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouChina
| | - Youyou Yan
- Translational Medicine Research CenterAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouChina
- Affiliated Hangzhou First People's HospitalZhejiang Chinese Medical UniversityHangzhouChina
| | - Hong Jiang
- Department of Thoracic SurgeryAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouChina
| | - Runlei Hu
- Department of Thoracic SurgeryAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouChina
| | | | | | - Jianguo Feng
- Cancer Research InstituteZhejiang Cancer HospitalHangzhouChina
| | - Nengming Lin
- Translational Medicine Research CenterAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouChina
- Affiliated Hangzhou First People's HospitalZhejiang Chinese Medical UniversityHangzhouChina
| |
Collapse
|
8
|
Cho HD, Gu IA, Won YS, Moon KD, Park KH, Seo KI. Auriculasin sensitizes primary prostate cancer cells to TRAIL-mediated apoptosis through up-regulation of the DR5-dependent pathway. Food Chem Toxicol 2019; 126:223-232. [PMID: 30817944 DOI: 10.1016/j.fct.2019.02.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 02/07/2019] [Accepted: 02/19/2019] [Indexed: 01/09/2023]
Abstract
Primary prostate cancer cells frequently develop resistance toward chemotherapy as well as most chemotherapeutics have been reported to induce undesirable cytotoxicity in normal cells. In this study, we performed sensitizing activity analysis of auriculasin (AC) to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in RC-58T/h/SA#4 primary prostate cancer cells without significant cytotoxicity in RWPE-1 prostate epithelial cells. Combined treatment with AC and TRAIL at optimal concentrations resulted in tumor-specific apoptotic cell death in RC-58T/h/SA#4 cells, characterized by DNA fragmentation, accumulation of apoptotic cell population, and nuclear condensation. Compared to single treatment with AC or TRAIL, co-treatment with AC and TRAIL significantly increased expression of Bax, cleaved PARP, AIF, endo G, and cytochrome c but decreased expression of phosphorylation of AKT and mammalian target of rapamycin (mTOR), phosphoinositide 3-kinase (PI3K), Bcl-2 and caspases-9, -8, -3, and -10. The sensitizing effect of AC to TRAIL was well correlated with inhibition of death receptor 5 (DR5) CHOP, and p53 expression. Moreover, pre-treatment with a chimeric blocking antibody for DR5 effectively reduced AC-TRAIL-induced cell death and apoptosis-related protein expression. These results suggest that non-toxic concentrations of AC sensitize TRAIL-resistant primary prostate cancer cells to TRAIL-mediated apoptosis via up-regulation of DR5 and downstream signaling pathways.
Collapse
Affiliation(s)
- Hyun-Dong Cho
- Department of Food Science and Technology, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - In-Ah Gu
- Department of Food Science, University of Arkansas, AR, 72704, USA
| | - Yeong-Seon Won
- Institute of Agriculture Life Science, Dong-A University, Busan, 49315, Republic of Korea
| | - Kwang-Deog Moon
- Department of Food Science and Technology, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Ki-Hun Park
- Division of Applied Life Science (BK21 plus), IALS, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Kwon-Il Seo
- Department of Biotechnology, Dong-A University, Busan, 49315, Republic of Korea.
| |
Collapse
|
9
|
Wang T, Cheng C, Peng L, Gao M, Xi M, Rousseaux S, Khochbin S, Wang J, Mi J. Combination of arsenic trioxide and Dasatinib: a new strategy to treat Philadelphia chromosome-positive acute lymphoblastic leukaemia. J Cell Mol Med 2017; 22:1614-1626. [PMID: 29266867 PMCID: PMC5824394 DOI: 10.1111/jcmm.13436] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have significantly improved the prognosis of Philadelphia chromosome-positive acute lymphoblastic leukaemia (Ph+ ALL), one of the most common and aggressive forms of haematological malignancies. However, TKI resistance has remained an unsolved issue. In this study, we investigate the impact of adding arsenic trioxide (ATO) on the action of Dasatinib, a second-generation TKI, in Ph+ ALL. We show that ATO cooperates with Dasatinib in both TKI-sensitive and resistant Ph+ ALL cell lines to increase apoptosis and we unravel the underlying mechanisms. Indeed, combining ATO and Dasatinib leads to severe cell apoptosis by activating the UPR apoptotic IRE1/JNK/PUMA axis, while neutralizing the UPR ATF4-dependent anti-apoptotic axis, activated by ATO alone. Additionally, ATO and Dasatinib in combination repress the expression of several genes, which we previously showed to be associated with shorter survival probability in ALL patients. Overall these data support the use of ATO in combination with Dasatinib as a novel therapeutic regimen for Ph+ ALL patients.
Collapse
Affiliation(s)
- Tao Wang
- State Key Laboratory for Medical Genomics and Department of Hematology, Shanghai Institute of Hematology, Collaborative Innovation Center of Systems Biomedicine, Pôle Sino-Français des Sciences du Vivant et Genomique, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunyan Cheng
- State Key Laboratory for Medical Genomics and Department of Hematology, Shanghai Institute of Hematology, Collaborative Innovation Center of Systems Biomedicine, Pôle Sino-Français des Sciences du Vivant et Genomique, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lijun Peng
- State Key Laboratory for Medical Genomics and Department of Hematology, Shanghai Institute of Hematology, Collaborative Innovation Center of Systems Biomedicine, Pôle Sino-Français des Sciences du Vivant et Genomique, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengqing Gao
- State Key Laboratory for Medical Genomics and Department of Hematology, Shanghai Institute of Hematology, Collaborative Innovation Center of Systems Biomedicine, Pôle Sino-Français des Sciences du Vivant et Genomique, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengping Xi
- State Key Laboratory for Medical Genomics and Department of Hematology, Shanghai Institute of Hematology, Collaborative Innovation Center of Systems Biomedicine, Pôle Sino-Français des Sciences du Vivant et Genomique, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sophie Rousseaux
- CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences, Université Grenoble-Alpes, La Tronche, France
| | - Saadi Khochbin
- CNRS UMR 5309, INSERM U1209, Institute for Advanced Biosciences, Université Grenoble-Alpes, La Tronche, France
| | - Jin Wang
- State Key Laboratory for Medical Genomics and Department of Hematology, Shanghai Institute of Hematology, Collaborative Innovation Center of Systems Biomedicine, Pôle Sino-Français des Sciences du Vivant et Genomique, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianqing Mi
- State Key Laboratory for Medical Genomics and Department of Hematology, Shanghai Institute of Hematology, Collaborative Innovation Center of Systems Biomedicine, Pôle Sino-Français des Sciences du Vivant et Genomique, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|