1
|
Carr A, Coulter JA, Workman J, Fay J, Farrelly A, Eustace AJ, Bennie L, Grogan L, Breathnach O, Morris PG, McNamara DA, Cremona M, O'Neill BDP, Hennessy BT, Toomey S. Targeting the phosphatidylinositol-3-kinase (PI3K) and mitogen activated protein kinase (MAPK) signalling pathways to enhance chemoradiotherapy in locally advanced rectal cancer. Cancer Treat Res Commun 2025; 43:100926. [PMID: 40245445 DOI: 10.1016/j.ctarc.2025.100926] [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: 10/15/2024] [Revised: 03/04/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
Responses to neoadjuvant chemoradiotherapy for locally advanced rectal cancer are not uniform. The phosphatidylinositol-3 kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways are involved in tumorigenesis and treatment resistance in many cancers; therefore, targeting these pathways could enhance response to chemoradiotherapy. A panel of colorectal cancer (CRC) cell lines (n = 10) with varying PI3K and MAPK mutational backgrounds were treated with combinations of 5-Flourouracil (5-FU), radiation, the PI3K inhibitor copanlisib, and/or the MEK inhibitor refametinib, and their effects on proliferation in vitro were measured. BALB/c SCID mice were implanted with CRC cell lines representative of each mutational background, treated with copanlisib and/or chemoradiotherapy, and monitored for tumor growth. In vitro, PIK3CA mutated cell lines were most sensitive to copanlisib (IC50=28 nM) and KRAS mutated cell lines were most sensitive to refametinib (IC50 = 36 nM), while the combination of copanlisib and refametinib was synergistic in 9/10 cell lines tested. The addition of copanlisib to 5-FU chemoradiotherapy inhibited cell growth compared to 5-FU chemoradiotherapy alone, an effect that was most notable in LS-1034 (KRAS mutated) and Caco-2 (PIK3CA/KRAS wild-type) cell lines. In vivo copanlisib and 5-FU chemoradiotherapy reduced tumor growth in all xenograft models and increased overall survival in LS-1034 and Caco-2 xenografts. Our results suggest that activation of the kinase signalling pathway may modulate PI3K/MEK inhibitor responsiveness in colorectal cancer. Furthermore, the addition of copanlisib to 5-FU chemoradiotherapy resulted in an enhanced anti-proliferative cytotoxic effect compared to 5-FU chemoradiotherapy alone, regardless of the background mutational status, and supports further clinical development of this regimen.
Collapse
Affiliation(s)
- Aoife Carr
- Department of Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | | | - Julie Workman
- Department of Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Joanna Fay
- Department of Pathology, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Angela Farrelly
- Department of Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Alex J Eustace
- Department of Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Lindsey Bennie
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| | - Liam Grogan
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland; Beaumont RCSI Cancer Centre, Dublin, Ireland
| | - Oscar Breathnach
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland; Beaumont RCSI Cancer Centre, Dublin, Ireland
| | - Patrick G Morris
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland; Beaumont RCSI Cancer Centre, Dublin, Ireland
| | - Deborah A McNamara
- Beaumont RCSI Cancer Centre, Dublin, Ireland; Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Mattia Cremona
- Department of Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Brian D P O'Neill
- Beaumont RCSI Cancer Centre, Dublin, Ireland; St. Luke's Radiation Oncology Network, Dublin, Ireland
| | - Bryan T Hennessy
- Department of Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland; Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland; Beaumont RCSI Cancer Centre, Dublin, Ireland
| | - Sinead Toomey
- Department of Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland; Beaumont RCSI Cancer Centre, Dublin, Ireland.
| |
Collapse
|
2
|
Lukacsovich D, Zambare W, Wu C, Huang H, Zhang W, Kim MJ, Alvarez J, Bercz A, Paty PB, Romesser PB, Wang L, Smith JJ, Chen XS. Integrating Tumor and Organoid DNA Methylation Profiles Reveals Robust Predictors of Chemotherapy Response in Rectal Cancer. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.28.25322951. [PMID: 40093220 PMCID: PMC11908278 DOI: 10.1101/2025.02.28.25322951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Rectal cancer patients display heterogeneous responses to neoadjuvant treatment-including the intensive total neoadjuvant therapy (TNT)-and reliable biomarkers are lacking to guide which tumors will benefit most from these regimens. Here, we profiled DNA methylation in tumor tissue and matched patient-derived organoids (PDOs) from 18 rectal cancer cases (50 total samples), leveraging the Illumina MethylationEPIC array and quality control filters that retained 771,964 CpG sites. Analyses used linear models (for tissue-only or PDO-only) and a joint linear mixed-effects approach (accounting for patient-level random effects) to identify significant CpGs associated with log-transformed FOLFOX IC50. We found that PDOs faithfully recapitulate patient-tumor methylation patterns (Spearman's correlation >0.95 among replicate organoids), and the joint model uncovered 745 CpGs tied to FOLFOX sensitivity, many of which were missed in tissue-only analyses. Differentially methylated regions reinforced that broader epigenetic blocks near TSS or enhancer regions may modulate chemo-resistance, while pathway enrichment pinpointed focal adhesion, ECM-receptor interaction, calcium signaling, and folate metabolism as key processes. A methylation risk score derived from these CpGs significantly predicted progression-free survival in an independent colorectal cancer cohort (p=0.019), outperforming single-sample-based signatures. These findings suggest that combining methylation profiles from both tumors and PDOs can expose robust epigenetic drivers of therapy response, aiding the development of clinically actionable biomarkers for rectal cancer TNT.
Collapse
Affiliation(s)
- David Lukacsovich
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Radiation Oncology, Colorectal and Anal Cancer Service, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Wini Zambare
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Chao Wu
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Hanchen Huang
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Radiation Oncology, Colorectal and Anal Cancer Service, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Wei Zhang
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Min Jung Kim
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Janet Alvarez
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Aron Bercz
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Philip B Paty
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Paul B Romesser
- Department of Radiation Oncology, Colorectal and Anal Cancer Service, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Lily Wang
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - J Joshua Smith
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - X Steven Chen
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| |
Collapse
|
3
|
Mousavikia SN, Darvish L, Firouzjaei AA, Toossi MTB, Azimian H. PI3K/AKT/mTOR Targeting in Colorectal Cancer Radiotherapy: A Systematic Review. J Gastrointest Cancer 2025; 56:52. [PMID: 39849185 DOI: 10.1007/s12029-024-01160-1] [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] [Accepted: 12/11/2024] [Indexed: 01/25/2025]
Abstract
BACKGROUND Radioresistance is a major challenge in the treatment of patients with colorectal cancer (CRC) and impairs the efficacy of radiotherapy. The PI3K/AKT/mTOR signaling pathway plays a critical role in CRC and contributes to the development of radioresistance. Accordingly, targeting this signaling pathway may be a promising strategy to improve oncotherapy. METHODS We performed a systematic search of Scopus, PubMed, Web of Science, Embase, and Medline databases. We included articles that investigated the effects of PI3K/AKT/mTOR pathway inhibitors on improving the efficacy of radiotherapy. RESULT Of the 32 articles included in our review, 27 were preclinical studies and 5 were clinical trials. We examined the effects of various signaling pathway inhibitors in combination with radiotherapy. While the efficacy of these therapies when used alone is limited, their combination is associated with reduced survival, induction of apoptosis, and cell cycle arrest, which may increase radiosensitivity. Despite the limited number of studies, this combination therapy has shown favorable treatment outcomes in patients with CRC. CONCLUSION PI3K/AKT/mTOR is a critical signaling pathway for cancer cell survival. By inhibiting this pathway, we can increase the efficacy of radiotherapy. These results provide valuable insights for the further development of research and clinical practice in the treatment of colorectal cancer.
Collapse
Affiliation(s)
- S N Mousavikia
- Medical Physics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - L Darvish
- Department of Radiology, Faculty of Paramedicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Mother and Child Welfare Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - A A Firouzjaei
- Bioinformatics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - M T Bahreyni Toossi
- Medical Physics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - H Azimian
- Medical Physics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Tufail M, Wan WD, Jiang C, Li N. Targeting PI3K/AKT/mTOR signaling to overcome drug resistance in cancer. Chem Biol Interact 2024; 396:111055. [PMID: 38763348 DOI: 10.1016/j.cbi.2024.111055] [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: 03/27/2024] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 05/21/2024]
Abstract
This review comprehensively explores the challenge of drug resistance in cancer by focusing on the pivotal PI3K/AKT/mTOR pathway, elucidating its role in oncogenesis and resistance mechanisms across various cancer types. It meticulously examines the diverse mechanisms underlying resistance, including genetic mutations, feedback loops, and microenvironmental factors, while also discussing the associated resistance patterns. Evaluating current therapeutic strategies targeting this pathway, the article highlights the hurdles encountered in drug development and clinical trials. Innovative approaches to overcome resistance, such as combination therapies and precision medicine, are critically analyzed, alongside discussions on emerging therapies like immunotherapy and molecularly targeted agents. Overall, this comprehensive review not only sheds light on the complexities of resistance in cancer but also provides a roadmap for advancing cancer treatment.
Collapse
Affiliation(s)
- Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Dong Wan
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Canhua Jiang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China; Institute of Oral Precancerous Lesions, Central South University, Changsha, China; Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ning Li
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China; Institute of Oral Precancerous Lesions, Central South University, Changsha, China; Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
5
|
Zhang H, Li X, Sun W, Qin H, Li H, Yan H, Wang H, Zhang X, Zhang S, Wang H. PTEN and P-4E-BP1 might be associated with postoperative recurrence of rectal cancer patients undergoing concurrent radiochemotherapy. BMC Cancer 2024; 24:582. [PMID: 38741069 PMCID: PMC11089754 DOI: 10.1186/s12885-024-12339-x] [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: 10/10/2023] [Accepted: 05/03/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Local recurrence after surgery and radiochemotherapy seriously affects the prognosis of locally advanced rectal cancer (LARC) patients. Studies on molecular markers related to the radiochemotherapy sensitivity of cancers have been widely carried out, which might provide valued information for clinicians to carry out individual treatment. AIM To find potential biomarkers of tumors for predicting postoperative recurrence. METHODS In this study, LARC patients undergoing surgery and concurrent radiochemotherapy were enrolled. We focused on clinicopathological factors and PTEN, SIRT1, p-4E-BP1, and pS6 protein expression assessed by immunohistochemistry in 73 rectal cancer patients with local recurrence and 76 patients without local recurrence. RESULTS The expression of PTEN was higher, while the expression of p-4E-BP1 was lower in patients without local recurrence than in patients with local recurrence. Moreover, TNM stage, lymphatic vessel invasion (LVI), PTEN and p-4E-BP1 might be independent risk factors for local recurrence after LARC surgery combined with concurrent radiochemotherapy. CONCLUSIONS This study suggests that PTEN and p-4E-BP1 might be potential biomarkers for prognostic prediction and therapeutic targets for LARC.
Collapse
Affiliation(s)
- Heng Zhang
- Department of Oncology, Tianjin Union Medical Center, Nankai University, No.190 Jieyuan Road, Hongqiao District, Tianjin, 300121, P. R. China
| | - Xiaofan Li
- Department of Oncology, People's Hospital of Rongcheng, Shandong, Rongcheng, P. R. China
| | - Wanjun Sun
- Department of Oncology, Tianjin Union Medical Center, Nankai University, No.190 Jieyuan Road, Hongqiao District, Tianjin, 300121, P. R. China
| | - Haoren Qin
- Department of Oncology, Tianjin Union Medical Center, Nankai University, No.190 Jieyuan Road, Hongqiao District, Tianjin, 300121, P. R. China
| | - Haipeng Li
- Department of Oncology, Tianjin Union Medical Center, Nankai University, No.190 Jieyuan Road, Hongqiao District, Tianjin, 300121, P. R. China
| | - Hao Yan
- Department of Oncology, Tianjin Union Medical Center, Nankai University, No.190 Jieyuan Road, Hongqiao District, Tianjin, 300121, P. R. China
| | - Huaqing Wang
- Department of Oncology, Tianjin Union Medical Center, Nankai University, No.190 Jieyuan Road, Hongqiao District, Tianjin, 300121, P. R. China
| | - Xipeng Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, Nankai University, Tianjin, P. R. China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Nankai University, Tianjin, P. R. China
| | - Hui Wang
- Department of Oncology, Tianjin Union Medical Center, Nankai University, No.190 Jieyuan Road, Hongqiao District, Tianjin, 300121, P. R. China.
| |
Collapse
|
6
|
Smithson M, Diffalha SA, Irwin RK, Williams G, McLeod MC, Somasundaram V, Bellis SL, Hardiman KM. ST6GAL1 is associated with poor response to chemoradiation in rectal cancer. Neoplasia 2024; 51:100984. [PMID: 38467087 PMCID: PMC11026834 DOI: 10.1016/j.neo.2024.100984] [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: 11/06/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/13/2024]
Abstract
INTRODUCTION Colorectal cancer is the third most common cause of cancer death. Rectal cancer makes up a third of all colorectal cases. Treatment for locally advanced rectal cancer includes chemoradiation followed by surgery. We have previously identified ST6GAL1 as a cause of resistance to chemoradiation in vitro and hypothesized that it would be correlated with poor response in human derived models and human tissues. METHODS Five organoid models were created from primary human rectal cancers and ST6GAL1 was knocked down via lentivirus transduction in one model. ST6GAL1 and Cleaved Caspase-3 (CC3) were assessed after chemoradiation via immunostaining. A tissue microarray (TMA) was created from twenty-six patients who underwent chemoradiation and had pre- and post-treatment specimens of rectal adenocarcinoma available at our institution. Immunohistochemistry was performed for ST6GAL1 and percent positive cancer cell staining was assessed and correlation with pathological grade of response was measured. RESULTS Organoid models were treated with chemoradiation and both ST6GAL1 mRNA and protein significantly increased after treatment. The organoid model targeted with ST6GAL1 knockdown was found to have increased CC3 after treatment. In the tissue microarray, 42 percent of patient samples had an increase in percent tumor cell staining for ST6GAL1 after treatment. Post-treatment percent staining was associated with a worse grade of treatment response (p = 0.01) and increased staining post-treatment compared to pre-treatment was also associated with a worse response (p = 0.01). CONCLUSION ST6GAL1 is associated with resistance to treatment in human rectal cancer and knockdown in an organoid model abrogated resistance to apoptosis caused by chemoradiation.
Collapse
Affiliation(s)
- Mary Smithson
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Al 35294, USA
| | - Sameer Al Diffalha
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Al 35294, USA
| | - Regina K Irwin
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Al 35294, USA
| | - Gregory Williams
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Al 35294, USA
| | - M Chandler McLeod
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Al 35294, USA
| | - Vivek Somasundaram
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Al 35294, USA
| | - Susan L Bellis
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Al 35294, USA
| | - Karin M Hardiman
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Al 35294, USA; Department of Surgery, Birmingham Veterans Affairs Medical Center, Birmingham, Al 35294, USA.
| |
Collapse
|
7
|
Pennel K, Dutton L, Melissourgou-Syka L, Roxburgh C, Birch J, Edwards J. Novel radiation and targeted therapy combinations for improving rectal cancer outcomes. Expert Rev Mol Med 2024; 26:e14. [PMID: 38623751 PMCID: PMC11140547 DOI: 10.1017/erm.2024.15] [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: 06/07/2023] [Revised: 01/29/2024] [Accepted: 03/22/2024] [Indexed: 04/17/2024]
Abstract
Neoadjuvant radiotherapy (RT) is commonly used as standard treatment for rectal cancer. However, response rates are variable and survival outcomes remain poor, highlighting the need to develop new therapeutic strategies. Research is focused on identifying novel methods for sensitising rectal tumours to RT to enhance responses and improve patient outcomes. This can be achieved through harnessing tumour promoting effects of radiation or preventing development of radio-resistance in cancer cells. Many of the approaches being investigated involve targeting the recently published new dimensions of cancer hallmarks. This review article will discuss key radiation and targeted therapy combination strategies being investigated in the rectal cancer setting, with a focus on exploitation of mechanisms which target the hallmarks of cancer.
Collapse
Affiliation(s)
- Kathryn Pennel
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
| | - Louise Dutton
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
| | - Lydia Melissourgou-Syka
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
- CRUK Scotland Institute, Glasgow, G611BD, UK
| | - Campbell Roxburgh
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
- Academic Unit of Surgery, Glasgow Royal Infirmary, University of Glasgow, Glasgow, G4 0SF, UK
| | - Joanna Birch
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
| | - Joanne Edwards
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
| |
Collapse
|
8
|
Wragg JW, White PL, Hadzhiev Y, Wanigasooriya K, Stodolna A, Tee L, Barros-Silva JD, Beggs AD, Müller F. Intra-promoter switch of transcription initiation sites in proliferation signaling-dependent RNA metabolism. Nat Struct Mol Biol 2023; 30:1970-1984. [PMID: 37996663 PMCID: PMC10716046 DOI: 10.1038/s41594-023-01156-8] [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: 01/30/2023] [Accepted: 10/19/2023] [Indexed: 11/25/2023]
Abstract
Global changes in transcriptional regulation and RNA metabolism are crucial features of cancer development. However, little is known about the role of the core promoter in defining transcript identity and post-transcriptional fates, a potentially crucial layer of transcriptional regulation in cancer. In this study, we use CAGE-seq analysis to uncover widespread use of dual-initiation promoters in which non-canonical, first-base-cytosine (C) transcription initiation occurs alongside first-base-purine initiation across 59 human cancers and healthy tissues. C-initiation is often followed by a 5' terminal oligopyrimidine (5'TOP) sequence, dramatically increasing the range of genes potentially subjected to 5'TOP-associated post-transcriptional regulation. We show selective, dynamic switching between purine and C-initiation site usage, indicating transcription initiation-level regulation in cancers. We additionally detail global metabolic changes in C-initiation transcripts that mark differentiation status, proliferative capacity, radiosensitivity, and response to irradiation and to PI3K-Akt-mTOR and DNA damage pathway-targeted radiosensitization therapies in colorectal cancer organoids and cancer cell lines and tissues.
Collapse
Affiliation(s)
- Joseph W Wragg
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| | - Paige-Louise White
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Yavor Hadzhiev
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Kasun Wanigasooriya
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Department of Surgery, University Hospitals Birmingham National Health Service (NHS) Foundation Trust, Birmingham, UK
| | - Agata Stodolna
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Louise Tee
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Joao D Barros-Silva
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Andrew D Beggs
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
- Department of Surgery, University Hospitals Birmingham National Health Service (NHS) Foundation Trust, Birmingham, UK.
| | - Ferenc Müller
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
9
|
Deng H, Chen Y, Wang L, Zhang Y, Hang Q, Li P, Zhang P, Ji J, Song H, Chen M, Jin Y. PI3K/mTOR inhibitors promote G6PD autophagic degradation and exacerbate oxidative stress damage to radiosensitize small cell lung cancer. Cell Death Dis 2023; 14:652. [PMID: 37802999 PMCID: PMC10558571 DOI: 10.1038/s41419-023-06171-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/12/2023] [Accepted: 09/21/2023] [Indexed: 10/08/2023]
Abstract
Our previous study revealed that PI3K/AKT/mTOR signaling was associated with SCLC radioresistance. SBC2 cells were used as primary radioresistance models, while H446 cells were continuously exposed to ionizing radiation (IR) to develop acquired radioresistance. Cell viability and apoptosis assays were used to investigate synergistic effects of BEZ235/GSK2126458 and IR in vitro, while immunoblotting, metabolite quantitative analysis and bioinformatic analyses were utilized to explore the underlying mechanism. Both genetically engineered mouse models (GEMM) and subcutaneous tumor models were used to confirm the synergistic effect in vivo. Key molecules of PI3K/AKT/mTOR signaling were upregulated after IR, which was correlated with primary radioresistance, and they were more expressed in acquired radioresistant cells. BEZ235/GSK2126458 effectively enhanced the cytotoxic effects of IR. BEZ235/GSK2126458 plus IR elevated γ-H2AX and p-Nrf2 expression, suggesting DNA and oxidative stress damage were intensified. Mechanistically, BEZ235/GSK2126458 plus IR significantly reduced the expression of G6PD protein, the rate-limiting enzyme of the pentose phosphate pathway (PPP). In detail, PI3K/mTOR inhibitors reinforced interaction between G6PD and HSPA8/HSC70, and G6PD was degraded by chaperone-mediated autophagy processes. Their metabolites (NADPH and R-5P) were decreased, and ROS levels were indirectly elevated, both of which exacerbated cell death. PI3K/AKT/mTOR signaling activator, insulin, enhanced SCLC radioresistance, while the synergistic effect of BEZ235/GSK2126458 and IR can be attenuated by N-acetylcysteine, and enhanced by 6-amino niacinamide. GEMM and allograft transplantation assays further confirmed their synergistic effect in vivo. This study provided insights into the connection between PI3K/AKT/mTOR signaling and the PPP underlying radioresistance and provided evidence of mechanisms supporting PI3K/mTOR inhibitors as possible therapeutic strategies to abrogate SCLC radioresistance.
Collapse
Affiliation(s)
- Huan Deng
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yamei Chen
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200240, China
| | - Li Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yibi Zhang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 331800, China
| | - Qingqing Hang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Peijing Li
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Peng Zhang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Jing Ji
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Hai Song
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| | - Ming Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
- United Laboratory of Frontier Radiotherapy Technology of Sun Yat-sen University & Chinese Academy of Sciences Ion Medical Technology Co., Ltd, Guangzhou, China.
| | - Ying Jin
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China.
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, 310022, China.
| |
Collapse
|
10
|
Fisch D, Pfleiderer MM, Anastasakou E, Mackie GM, Wendt F, Liu X, Clough B, Lara-Reyna S, Encheva V, Snijders AP, Bando H, Yamamoto M, Beggs AD, Mercer J, Shenoy AR, Wollscheid B, Maslowski KM, Galej WP, Frickel EM. PIM1 controls GBP1 activity to limit self-damage and to guard against pathogen infection. Science 2023; 382:eadg2253. [PMID: 37797010 PMCID: PMC7615196 DOI: 10.1126/science.adg2253] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 08/23/2023] [Indexed: 10/07/2023]
Abstract
Disruption of cellular activities by pathogen virulence factors can trigger innate immune responses. Interferon-γ (IFN-γ)-inducible antimicrobial factors, such as the guanylate binding proteins (GBPs), promote cell-intrinsic defense by attacking intracellular pathogens and by inducing programmed cell death. Working in human macrophages, we discovered that GBP1 expression in the absence of IFN-γ killed the cells and induced Golgi fragmentation. IFN-γ exposure improved macrophage survival through the activity of the kinase PIM1. PIM1 phosphorylated GBP1, leading to its sequestration by 14-3-3σ, which thereby prevented GBP1 membrane association. During Toxoplasma gondii infection, the virulence protein TgIST interfered with IFN-γ signaling and depleted PIM1, thereby increasing GBP1 activity. Although infected cells can restrain pathogens in a GBP1-dependent manner, this mechanism can protect uninfected bystander cells. Thus, PIM1 can provide a bait for pathogen virulence factors, guarding the integrity of IFN-γ signaling.
Collapse
Affiliation(s)
- Daniel Fisch
- Host-Toxoplasma Interaction Laboratory, The Francis Crick Institute, London, UK
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, UK
| | - Moritz M Pfleiderer
- European Molecular Biology Laboratory, 71 Avenue des Martyrs, Grenoble, France
| | - Eleni Anastasakou
- European Molecular Biology Laboratory, 71 Avenue des Martyrs, Grenoble, France
| | - Gillian M Mackie
- Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, UK
| | - Fabian Wendt
- Department of Health Sciences and Technology (D-HEST), ETH Zurich, Institute of Translational Medicine (ITM), Zurich, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Xiangyang Liu
- European Molecular Biology Laboratory, 71 Avenue des Martyrs, Grenoble, France
| | - Barbara Clough
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, UK
| | - Samuel Lara-Reyna
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, UK
| | - Vesela Encheva
- Mass Spectrometry and Proteomics Platform, The Francis Crick Institute, London, UK
| | - Ambrosius P Snijders
- Mass Spectrometry and Proteomics Platform, The Francis Crick Institute, London, UK
- Bruker Nederland BV
| | - Hironori Bando
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Andrew D Beggs
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, UK
| | - Jason Mercer
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, UK
| | - Avinash R Shenoy
- MRC Centre for Molecular Bacteriology & Infection, Department of Infectious Disease, Imperial College London, London, UK
- The Francis Crick Institute, London, UK
| | - Bernd Wollscheid
- Department of Health Sciences and Technology (D-HEST), ETH Zurich, Institute of Translational Medicine (ITM), Zurich, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Kendle M Maslowski
- Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, UK
- Cancer Research UK Beatson Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Wojtek P Galej
- European Molecular Biology Laboratory, 71 Avenue des Martyrs, Grenoble, France
| | - Eva-Maria Frickel
- Host-Toxoplasma Interaction Laboratory, The Francis Crick Institute, London, UK
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, UK
| |
Collapse
|
11
|
Landon‐Brace N, Li NT, McGuigan AP. Exploring New Dimensions of Tumor Heterogeneity: The Application of Single Cell Analysis to Organoid-Based 3D In Vitro Models. Adv Healthc Mater 2023; 12:e2300903. [PMID: 37589373 PMCID: PMC11468421 DOI: 10.1002/adhm.202300903] [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: 03/21/2023] [Revised: 06/28/2023] [Indexed: 08/18/2023]
Abstract
Modeling the heterogeneity of the tumor microenvironment (TME) in vitro is essential to investigating fundamental cancer biology and developing novel treatment strategies that holistically address the factors affecting tumor progression and therapeutic response. Thus, the development of new tools for both in vitro modeling, such as patient-derived organoids (PDOs) and complex 3D in vitro models, and single cell omics analysis, such as single-cell RNA-sequencing, represents a new frontier for investigating tumor heterogeneity. Specifically, the integration of PDO-based 3D in vitro models and single cell analysis offers a unique opportunity to explore the intersecting effects of interpatient, microenvironmental, and tumor cell heterogeneity on cell phenotypes in the TME. In this review, the current use of PDOs in complex 3D in vitro models of the TME is discussed and the emerging directions in the development of these models are highlighted. Next, work that has successfully applied single cell analysis to PDO-based models is examined and important experimental considerations are identified for this approach. Finally, open questions are highlighted that may be amenable to exploration using the integration of PDO-based models and single cell analysis. Ultimately, such investigations may facilitate the identification of novel therapeutic targets for cancer that address the significant influence of tumor-TME interactions.
Collapse
Affiliation(s)
- Natalie Landon‐Brace
- Institute of Biomedical EngineeringUniversity of Toronto200 College StreetTorontoM5S3E5Canada
| | - Nancy T. Li
- Department of Chemical Engineering and Applied ChemistryUniversity of Toronto200 College StTorontoM5S3E5Canada
| | - Alison P. McGuigan
- Department of Chemical Engineering and Applied ChemistryInstitute of Biomedical EngineeringUniversity of Toronto200 College StTorontoM5S3E5Canada
| |
Collapse
|
12
|
Lin H, Wang Y, Cheng C, Qian Y, Hao J, Zhang Z, Sheng W, Song L, Deng CX, Zhao B, Cao J, Wang L, Wang L, Liang L, Chen WK, Yu C, Sun Z, Yang Y, Wang C, Zhang Y, Li Q, Li K, Ma A, Zhao T, Chen YG, Hua G. Standard: Human intestinal cancer organoids. CELL REGENERATION (LONDON, ENGLAND) 2023; 12:24. [PMID: 37378693 DOI: 10.1186/s13619-023-00167-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Intestinal cancer is one of the most frequent and lethal types of cancer. Modeling intestinal cancer using organoids has emerged in the last decade. Human intestinal cancer organoids are physiologically relevant in vitro models, which provides an unprecedented opportunity for fundamental and applied research in colorectal cancer. "Human intestinal cancer organoids" is the first set of guidelines on human intestinal organoids in China, jointly drafted and agreed by the experts from the Chinese Society for Cell Biology and its branch society: the Chinese Society for Stem Cell Research. This standard specifies terms and definitions, technical requirements, test methods for human intestinal cancer organoids, which apply to the production and quality control during the process of manufacturing and testing of human intestinal cancer organoids. It was released by the Chinese Society for Cell Biology on 24 September 2022. We hope that the publication of this standard will guide institutional establishment, acceptance and execution of proper practocal protocols, and accelerate the international standardization of human intestinal cancer organoids for clinical development and therapeutic applications.
Collapse
Affiliation(s)
- Hanqing Lin
- D1Med Technology (Shanghai) Inc, Shanghai, 201802, China
| | - Yalong Wang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Guangzhou Laboratory, Guangzhou, 510005, China
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Guangzhou Hua Yi Regeneration Technology Co., Ltd, Huangpu District, Guangzhou, 510700, China
| | - Chunyan Cheng
- D1Med Technology (Shanghai) Inc, Shanghai, 201802, China
| | - Yuxin Qian
- D1Med Technology (Shanghai) Inc, Shanghai, 201802, China
| | - Jie Hao
- National Stem Cell Resource Center, State Key Laboratory of Stem Cell and Reproductive Biology, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Zhen Zhang
- Department of Radiation Oncology and Cancer Institute, Fudan University Shanghai Cancer Center Fudan University, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Weiqi Sheng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Linhong Song
- Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, 610072, China
| | - Chu-Xia Deng
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau, 999078, SAR, China
| | - Bing Zhao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Jiani Cao
- National Stem Cell Resource Center, State Key Laboratory of Stem Cell and Reproductive Biology, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Lei Wang
- National Stem Cell Resource Center, State Key Laboratory of Stem Cell and Reproductive Biology, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Liu Wang
- National Stem Cell Resource Center, State Key Laboratory of Stem Cell and Reproductive Biology, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Lingmin Liang
- National Stem Cell Resource Center, State Key Laboratory of Stem Cell and Reproductive Biology, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Wenli Kelly Chen
- China Innovation Center of Roche, Li Shi Zhen Road, Pudong, Shanghai, 201203, China
| | - Chunping Yu
- Eli Lilly and Company, Pudong, Shanghai, 201203, China
| | - Zhijian Sun
- K2 Oncology Co., Ltd, KeChuang Street, Beijing, 100176, China
| | | | - Changlin Wang
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
- China National Institute of Standardization, Beijing, 100191, China
| | - Yong Zhang
- Chinese Society for Stem Cell Research, Shanghai, 200032, China
- HHLIFE Co., Inc, Shenzhen, 518040, China
| | - Qiyuan Li
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
- China National GeneBank, Shenzhen, 518000, China
| | - Ka Li
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
- Chinese Society for Stem Cell Research, Shanghai, 200032, China
| | - Aijin Ma
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, 200433, China.
- Beijing Technology and Business University, Beijing, 100048, China.
| | - Tongbiao Zhao
- National Stem Cell Resource Center, State Key Laboratory of Stem Cell and Reproductive Biology, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, 200433, China.
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Guangzhou Laboratory, Guangzhou, 510005, China.
- School of Basic Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| | - Guoqiang Hua
- D1Med Technology (Shanghai) Inc, Shanghai, 201802, China.
- Department of Radiation Oncology and Cancer Institute, Fudan University Shanghai Cancer Center Fudan University, Shanghai, 200032, China.
| |
Collapse
|