1
|
Conceição CJF, Moe E, Ribeiro PA, Raposo M. PARP1: A comprehensive review of its mechanisms, therapeutic implications and emerging cancer treatments. Biochim Biophys Acta Rev Cancer 2025; 1880:189282. [PMID: 39947443 DOI: 10.1016/j.bbcan.2025.189282] [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: 05/22/2024] [Revised: 01/28/2025] [Accepted: 02/04/2025] [Indexed: 02/21/2025]
Abstract
The Poly (ADP-ribose) polymerase-1 (PARP1) enzyme is involved in several signalling pathways related to homologous repair (HR), base excision repair (BER), and non-homologous end joining (NHEJ). Studies demonstrated that the deregulation of PARP1 function and control mechanisms can lead to cancer emergence. On the other side, PARP1 can be a therapeutic target to maximize cancer treatment. This is done by molecules that can modulate radiation effects, such as DNA repair inhibitors (PARPi). With this approach, tumour cell viability can be undermined by targeting DNA repair mechanisms. Thus, treatment using PARPi represents a new era for cancer therapy, and even new horizons can be attained by coupling these molecules with a nano-delivery system. For this, drug delivery systems such as liposomes encompass all the required features due to its excellent biocompatibility, biodegradability, and low toxicity. This review presents a comprehensive overview of PARP1 biological features and mechanisms, its role in cancer development, therapeutic implications, and emerging cancer treatments by PARPi-mediated therapies. Although there are a vast number of studies regarding PARP1 biological function, some PARP1 mechanisms are not clear yet, and full-length PARP1 structure is missing. Nevertheless, literature reports demonstrate already the high usefulness and vast possibilities offered by combined PARPi cancer therapy.
Collapse
Affiliation(s)
- Carlota J F Conceição
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal.
| | - Elin Moe
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal; Department of Chemistry, UiT-The Arctic University of Norway, N-9037 Tromsø, Norway.
| | - Paulo A Ribeiro
- Laboratory of Instrumentation, Biomedical Engineering and Radiation Physics (LIBPhys-UNL), Department of Physics, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal.
| | - Maria Raposo
- Laboratory of Instrumentation, Biomedical Engineering and Radiation Physics (LIBPhys-UNL), Department of Physics, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal.
| |
Collapse
|
2
|
Smith HL, Willmore E, Prendergast L, Curtin NJ. ATR, CHK1 and WEE1 inhibitors cause homologous recombination repair deficiency to induce synthetic lethality with PARP inhibitors. Br J Cancer 2024; 131:905-917. [PMID: 38965423 PMCID: PMC11369084 DOI: 10.1038/s41416-024-02745-0] [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: 08/03/2023] [Revised: 05/24/2024] [Accepted: 05/31/2024] [Indexed: 07/06/2024] Open
Abstract
PURPOSE PARP inhibitors (PARPi) are effective in homologous recombination repair (HRR) defective (HRD) cancers. To (re)sensitise HRR proficient (HRP) tumours to PARPi combinations with other drugs are being explored. Our aim was to determine the mechanism underpinning the sensitisation to PARPi by inhibitors of cell cycle checkpoint kinases ATR, CHK1 and WEE1. EXPERIMENTAL DESIGN A panel of HRD and HRP cells (including matched BRCA1 or 2 mutant and corrected pairs) and ovarian cancer ascites cells were used. Rucaparib (PARPi) induced replication stress (RS) and HRR (immunofluorescence microscopy for γH2AX and RAD51 foci, respectively), cell cycle changes (flow cytometry), activation of ATR, CHK1 and WEE1 (Western Blot for pCHK1S345, pCHK1S296 and pCDK1Y15, respectively) and cytotoxicity (colony formation assay) was determined, followed by investigations of the impact on all of these parameters by inhibitors of ATR (VE-821, 1 µM), CHK1 (PF-477736, 50 nM) and WEE1 (MK-1775, 100 nM). RESULTS Rucaparib induced RS (3 to10-fold), S-phase accumulation (2-fold) and ATR, CHK1 and WEE1 activation (up to 3-fold), and VE-821, PF-477736 and MK-1775 inhibited their targets and abrogated these rucaparib-induced cell cycle changes in HRP and HRD cells. Rucaparib activated HRR in HRP cells only and was (60-1,000x) more cytotoxic to HRD cells. VE-821, PF-477736 and MK-1775 blocked HRR and sensitised HRP but not HRD cells and primary ovarian ascites to rucaparib. CONCLUSIONS Our data indicate that, rather than acting via abrogation of cell cycle checkpoints, ATR, CHK1 and WEE1 inhibitors cause an HRD phenotype and hence "induced synthetic lethality" with PARPi.
Collapse
Affiliation(s)
- Hannah L Smith
- Faculty of Medical Sciences, Newcastle University Centre for Cancer, Newcastle upon Tyne, NE1 7RU, UK.
| | - Elaine Willmore
- Faculty of Medical Sciences, Newcastle University Centre for Cancer, Newcastle upon Tyne, NE1 7RU, UK
| | - Lisa Prendergast
- Faculty of Medical Sciences, Newcastle University Centre for Cancer, Newcastle upon Tyne, NE1 7RU, UK
| | - Nicola J Curtin
- Faculty of Medical Sciences, Newcastle University Centre for Cancer, Newcastle upon Tyne, NE1 7RU, UK.
| |
Collapse
|
3
|
Derby S, Jackson MR, Williams K, Stobo J, Kelly C, Sweeting L, Shad S, Herbert C, Short SC, Williamson A, James A, Nowicki S, Bulbeck H, Chalmers AJ. Concurrent Olaparib and Radiation Therapy in Older Patients With Newly Diagnosed Glioblastoma: The Phase 1 Dose-Escalation PARADIGM Trial. Int J Radiat Oncol Biol Phys 2024; 118:1371-1378. [PMID: 38211641 DOI: 10.1016/j.ijrobp.2024.01.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/04/2024] [Accepted: 01/04/2024] [Indexed: 01/13/2024]
Abstract
PURPOSE Patients with glioblastoma who are older or have poor performance status (PS) experience particularly poor clinical outcomes. At the time of study initiation, these patients were treated with short-course radiation therapy (40 Gy in 15 fractions). Olaparib is an oral inhibitor of the DNA repair enzyme poly (ADP-ribose) polymerase (PARP) that is well tolerated as a single agent but exacerbates acute radiation toxicity in extracranial sites. Preclinical data predicted that PARP inhibitors would enhance radiosensitivity in glioblastoma without exacerbating adverse effects on the normal brain. METHODS AND MATERIALS Phase 1 of the PARADIGM trial was a 3+3 dose-escalation study testing olaparib in combination with radiation therapy (40 Gy 15 fractions) in patients with newly diagnosed glioblastoma who were unsuitable for radical treatment either because they were aged 70 years or older (World Health Organization PS 0-1) or aged 18 to 69 years with PS 2. The primary outcome was the recommended phase 2 dose of olaparib. Secondary endpoints included safety and tolerability, overall survival, and progression-free survival. Effects on cognitive function were assessed via the Mini Mental State Examination. RESULTS Of 16 eligible patients (56.25% male; median age, 71.5 years [range, 44-78]; 75% PS 0-1), 1 dose-limiting toxicity was reported (grade 3 agitation). Maximum tolerated dose was not reached and the recommended phase 2 dose was determined as 200 mg twice daily. Median overall survival and progression-free survival were 10.8 months (80% CI, 7.3-11.4) and 5.5 months (80% CI, 3.9-5.9), respectively. Mini Mental State Examination plots indicated that cognitive function was not adversely affected by the olaparib-radiation therapy combination. CONCLUSIONS Olaparib can be safely combined with hypofractionated brain radiation therapy and is well tolerated in patients unsuitable for radical chemoradiation. These results enabled initiation of a randomized phase 2 study and support future trials of PARP inhibitors in combination with radiation therapy for patients with brain tumors.
Collapse
Affiliation(s)
- Sarah Derby
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Mark R Jackson
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Karin Williams
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Jamie Stobo
- CRUK Glasgow Clinical Trials Unit, University of Glasgow, Glasgow, United Kingdom
| | - Caroline Kelly
- CRUK Glasgow Clinical Trials Unit, University of Glasgow, Glasgow, United Kingdom
| | - Lorna Sweeting
- CRUK Glasgow Clinical Trials Unit, University of Glasgow, Glasgow, United Kingdom
| | - Shumaila Shad
- CRUK Glasgow Clinical Trials Unit, University of Glasgow, Glasgow, United Kingdom
| | - Christopher Herbert
- University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | - Susan C Short
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | | | - Allan James
- Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Stefan Nowicki
- Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | | | - Anthony J Chalmers
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom; Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom.
| |
Collapse
|
4
|
Frey B, Borgmann K, Jost T, Greve B, Oertel M, Micke O, Eckert F. DNA as the main target in radiotherapy-a historical overview from first isolation to anti-tumour immune response. Strahlenther Onkol 2023; 199:1080-1090. [PMID: 37620671 DOI: 10.1007/s00066-023-02122-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 07/10/2023] [Indexed: 08/26/2023]
Abstract
DNA damage is one of the foremost mechanisms of irradiation at the biological level. After the first isolation of DNA by Friedrich Miescher in the 19th century, the structure of DNA was described by Watson and Crick. Several Nobel Prizes have been awarded for DNA-related discoveries. This review aims to describe the historical perspective of DNA in radiation biology. Over the decades, DNA damage has been identified and quantified after irradiation. Depending on the type of sensing, different proteins are involved in sensing DNA damage and repairing the damage, if possible. For double-strand breaks, the main repair mechanisms are non-homologous end joining and homologous recombination. Additional mechanisms are the Fanconi anaemia pathway and base excision repair. Different methods have been developed for the detection of DNA double-strand breaks. Several drugs have been developed that interfere with different DNA repair mechanisms, e.g., PARP inhibitors. These drugs have been established in the standard treatment of different tumour entities and are being applied in several clinical trials in combination with radiotherapy. Over the past decades, it has become apparent that DNA damage mechanisms are also directly linked to the immune response in tumours. For example, cytosolic DNA fragments activate the innate immune system via the cGAS STING pathway.
Collapse
Affiliation(s)
- Benjamin Frey
- Translational Radiation Biology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Kerstin Borgmann
- Laboratory of Radiobiology and Radiation Oncology, Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tina Jost
- Translational Radiation Biology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Burkhard Greve
- Department of Radiation Oncology, University Hospital Muenster, Muenster, Germany
| | - Michael Oertel
- Department of Radiation Oncology, University Hospital Muenster, Muenster, Germany
| | - Oliver Micke
- Department of Radiotherapy and Radiation Oncology, Franziskus Hospital Bielefeld, Kiskerst. 26, 33615, Bielefeld, Germany.
| | - Franziska Eckert
- Department of Radiation Oncology, AKH, Comprehensive Cancer Center Vienna, Medical University Vienna, Vienna, Austria
| |
Collapse
|
5
|
Vanderlinden A, Jones CG, Myers KN, Rominiyi O, Collis SJ. DNA damage response inhibitors enhance tumour treating fields (TTFields) potency in glioma stem-like cells. Br J Cancer 2023; 129:1829-1840. [PMID: 37777579 PMCID: PMC10667536 DOI: 10.1038/s41416-023-02454-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/02/2023] Open
Abstract
BACKGROUND High-grade gliomas are primary brain cancers with unacceptably low and persistent survival rates of 10-16 months for WHO grade 4 gliomas over the last 40 years, despite surgical resection and DNA-damaging chemo-radiotherapy. More recently, tumour-treating fields therapy (TTFields) has demonstrated modest survival benefit and been clinically approved in several countries. TTFields is thought to mediate anti-cancer activity by primarily disrupting mitosis. However, recent data suggest that TTFields may also attenuate DNA damage repair and replication fork dynamics, providing a potential platform for therapeutic combinations incorporating standard-of-care treatments and targeted DNA damage response inhibitors (DDRi). METHODS We have used patient-derived, typically resistant, glioma stem-like cells (GSCs) in combination with the previously validated preclinical Inovitro™ TTFields system together with a number of therapeutic DDRi. RESULTS We show that TTFields robustly activates PARP- and ATR-mediated DNA repair (including PARylation and CHK1 phosphorylation, respectively), whilst combining TTFields with PARP1 or ATR inhibitor treatment leads to significantly reduced clonogenic survival. The potency of each of these strategies is further enhanced by radiation treatment, leading to increased amounts of DNA damage with profound delay in DNA damage resolution. CONCLUSION To our knowledge, our findings represent the first report of TTFields applied with clinically approved or in-trial DDRi in GSC models and provides a basis for translational studies toward multimodal DDRi/TTFields-based therapeutic strategies for patients with these currently incurable tumours.
Collapse
Affiliation(s)
- Aurelie Vanderlinden
- Division of Clinical Medicine, The University of Sheffield, School of Medicine and Population Health, Sheffield, S10 2RX, UK
| | - Callum G Jones
- Division of Clinical Medicine, The University of Sheffield, School of Medicine and Population Health, Sheffield, S10 2RX, UK
| | - Katie N Myers
- Division of Clinical Medicine, The University of Sheffield, School of Medicine and Population Health, Sheffield, S10 2RX, UK
| | - Ola Rominiyi
- Division of Clinical Medicine, The University of Sheffield, School of Medicine and Population Health, Sheffield, S10 2RX, UK.
- Division of Neuroscience, The University of Sheffield, School of Medicine and Population Health, Sheffield, S10 2RX, UK.
- Department of Neurosurgery, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, S10 2JF, UK.
| | - Spencer J Collis
- Division of Clinical Medicine, The University of Sheffield, School of Medicine and Population Health, Sheffield, S10 2RX, UK.
| |
Collapse
|
7
|
Shen D, Luo J, Chen L, Ma W, Mao X, Zhang Y, Zheng J, Wang Y, Wan J, Wang S, Ouyang J, Yi H, Liu D, Huang W, Zhang W, Liu Z, McLeod HL, He Y. PARPi treatment enhances radiotherapy-induced ferroptosis and antitumor immune responses via the cGAS signaling pathway in colorectal cancer. Cancer Lett 2022; 550:215919. [PMID: 36116741 DOI: 10.1016/j.canlet.2022.215919] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/01/2022] [Accepted: 09/11/2022] [Indexed: 12/17/2022]
Abstract
In cancer cells, poly (ADP-ribose) polymerase (PARP)-1 and PARP2 initiate and regulate DNA repair pathways to protect against DNA damage and cell death caused by radiotherapy or chemotherapy. Radiotherapy and PARP inhibitors (PARPis) have been combined in clinical trials, but their action mechanisms remain unclear. Here, we show that activated by ionizing radiation (IR) generated dsDNA, cyclic GMP-AMP synthase (cGAS) signaling promoted regulated cell death, specifically ferroptosis, via the activating transcription factor 3 (ATF3)-solute carrier family 7 member 11 axis and the antitumor immune response via the interferon-β-CD8+ T cell pathway. Niraparib, a widely used PARPi, augmented cGAS-mediated ferroptosis and immune activation. In colorectal cancer models, cGAS knockdown (KD) compromised IR-induced ferroptosis via downregulation of ATF3 (key ferroptosis regulator) expression. cGAS depletion reversed IR-induced infiltration of CD8+ T or CD8+GZMB+ T cells in the cGAS KD group. Survival analysis of paired tumor samples before and after standard radiotherapy revealed that high expression levels of cGAS, ATF3, and PTGS2 and high density of CD8+ T cells resulted in a significantly high disease-free survival rate in patients with rectal cancer. Therefore, PARPi treatment increases the cytoplasmic accumulation of dsDNA caused by IR, triggering the cGAS signaling-mediated tumor control in cancer cell lines and mouse xenograft models.
Collapse
Affiliation(s)
- Dongya Shen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Pharmacogenetics, Central South University, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, China
| | - Jia Luo
- Department of Hepatobiliary and Intestinal Surgery, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Ling Chen
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenjuan Ma
- Department of Anesthesiology, Xiangya Hospital, Central South University, China
| | - Xiaoyuan Mao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Pharmacogenetics, Central South University, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, China
| | - Yu Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Pharmacogenetics, Central South University, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, China
| | - Juyan Zheng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Pharmacogenetics, Central South University, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, China
| | - Yang Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Pharmacogenetics, Central South University, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, China
| | - Jielin Wan
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Pharmacogenetics, Central South University, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, China
| | - Shiyu Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Pharmacogenetics, Central South University, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, China
| | - Jing Ouyang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Pharmacogenetics, Central South University, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, China
| | - Hanying Yi
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Pharmacogenetics, Central South University, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, China
| | - Dongbo Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Pharmacogenetics, Central South University, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, China
| | - Weihua Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Pharmacogenetics, Central South University, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Pharmacogenetics, Central South University, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, China; The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510060, China
| | - Zhaoqian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Pharmacogenetics, Central South University, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, China
| | - Howard L McLeod
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, China; Intermountain Precision Genomics, Intermountain Healthcare, St. George, UT, 84770, USA.
| | - Yijing He
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Pharmacogenetics, Central South University, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Central South University, China.
| |
Collapse
|