1
|
Yang Y, Zhang X, Yang Y, Gao P, Fan W, Zheng T, Yang W, Tang Y, Cai K. A two-pronged approach to inhibit ferroptosis of MSCs caused by the iron overload in postmenopausal osteoporosis and promote osseointegration of titanium implant. Bioact Mater 2024; 41:336-354. [PMID: 39161794 PMCID: PMC11331706 DOI: 10.1016/j.bioactmat.2024.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 07/02/2024] [Accepted: 07/15/2024] [Indexed: 08/21/2024] Open
Abstract
Postmenopausal osteoporosis (PMOP) is a prevalent condition among elderly women. After menopause, women exhibit decreased iron excretion, which is prone to osteoporosis. To design a specific titanium implant for PMOP, we first analyze miRNAs and DNA characteristics of postmenopausal patients with and without osteoporosis. The results indicate that iron overload disrupts iron homeostasis in the pathogenesis of PMOP. Further experiments confirm that iron overload can cause lipid peroxidation and ferroptosis of MSCs, thus breaking bone homeostasis. Based on the findings above, we have designed a novel Ti implant coated with nanospheres of caffeic acid (CA) and deferoxamine (DFO). CA can bind on the Ti surface through the two adjacent phenolic hydroxyls and polymerize into polycaffeic acid (PCA) dimer, as well as the PCA nanospheres with the repetitive 1,4-benzodioxan units. DFO was grafted with PCA through borate ester bonds. The experimental results showed that modified Ti can inhibit the ferroptosis of MSCs in the pathological environment of PMOP and promote osseointegration in two main ways. Firstly, DFO was released under high oxidative stress, chelating with excess iron and decreasing the labile iron pool in MSCs. Meanwhile, CA and DFO activated the KEAP1/NRF2/HMOX1 pathway in MSCs and reduced the level of intracellular lipid peroxidation. So, the ferroptosis of MSCs is inhibited by promoting the SLC7A11/GSH/GPX4 pathway. Furthermore, the remained CA coating on the Ti surface could reduce the extracellular oxidative stress and glutathione level. This study offers a novel inspiration for the specific design of Ti implants in the treatment of PMOP.
Collapse
Affiliation(s)
- Yulu Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Xianhui Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yao Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Pengfei Gao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Wuzhe Fan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Tao Zheng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Weihu Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yu Tang
- Orthopedics Department, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| |
Collapse
|
2
|
Hayashi M, Okazaki K, Papgiannakopoulos T, Motohashi H. The Complex Roles of Redox and Antioxidant Biology in Cancer. Cold Spring Harb Perspect Med 2024; 14:a041546. [PMID: 38772703 PMCID: PMC11529857 DOI: 10.1101/cshperspect.a041546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
Redox reactions control fundamental biochemical processes, including energy production, metabolism, respiration, detoxification, and signal transduction. Cancer cells, due to their generally active metabolism for sustained proliferation, produce high levels of reactive oxygen species (ROS) compared to normal cells and are equipped with antioxidant defense systems to counteract the detrimental effects of ROS to maintain redox homeostasis. The KEAP1-NRF2 system plays a major role in sensing and regulating endogenous antioxidant defenses in both normal and cancer cells, creating a bivalent contribution of NRF2 to cancer prevention and therapy. Cancer cells hijack the NRF2-dependent antioxidant program and exploit a very unique metabolism as a trade-off for enhanced antioxidant capacity. This work provides an overview of redox metabolism in cancer cells, highlighting the role of the KEAP1-NRF2 system, selenoproteins, sulfur metabolism, heme/iron metabolism, and antioxidants. Finally, we describe therapeutic approaches that can be leveraged to target redox metabolism in cancer.
Collapse
Affiliation(s)
- Makiko Hayashi
- Department of Pathology, New York University School of Medicine, New York, New York 10016, USA
| | - Keito Okazaki
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | | | - Hozumi Motohashi
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| |
Collapse
|
3
|
Lin YC, Ku CC, Wuputra K, Wu DC, Yokoyama KK. Vulnerability of Antioxidant Drug Therapies on Targeting the Nrf2-Trp53-Jdp2 Axis in Controlling Tumorigenesis. Cells 2024; 13:1648. [PMID: 39404411 PMCID: PMC11475825 DOI: 10.3390/cells13191648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024] Open
Abstract
Control of oxidation/antioxidation homeostasis is important for cellular protective functions, and disruption of the antioxidation balance by exogenous and endogenous ligands can lead to profound pathological consequences of cancerous commitment within cells. Although cancers are sensitive to antioxidation drugs, these drugs are sometimes associated with problems including tumor resistance or dose-limiting toxicity in host animals and patients. These problems are often caused by the imbalance between the levels of oxidative stress-induced reactive oxygen species (ROS) and the redox efficacy of antioxidants. Increased ROS levels, because of abnormal function, including metabolic abnormality and signaling aberrations, can promote tumorigenesis and the progression of malignancy, which are generated by genome mutations and activation of proto-oncogene signaling. This hypothesis is supported by various experiments showing that the balance of oxidative stress and redox control is important for cancer therapy. Although many antioxidant drugs exhibit therapeutic potential, there is a heterogeneity of antioxidation functions, including cell growth, cell survival, invasion abilities, and tumor formation, as well as the expression of marker genes including tumor suppressor proteins, cell cycle regulators, nuclear factor erythroid 2-related factor 2, and Jun dimerization protein 2; their effectiveness in cancer remains unproven. Here, we summarize the rationale for the use of antioxidative drugs in preclinical and clinical antioxidant therapy of cancer, and recent advances in this area using cancer cells and their organoids, including the targeting of ROS homeostasis.
Collapse
Affiliation(s)
- Ying-Chu Lin
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Chia-Chen Ku
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.K.); (K.W.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Kenly Wuputra
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.K.); (K.W.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Deng-Chyang Wu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Kazunari K. Yokoyama
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.K.); (K.W.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| |
Collapse
|
4
|
Zumsteg ZS, Sheth S, Jabbour SK, Patel KR, Kimple RJ, Williams TM, Xu-Welliver M, Torres-Saavedra PA, Monjazeb AM, Mayadev J, Finkelstein SE, Buatti JM, Patel SP, Lin SH. Challenges and opportunities for early phase clinical trials of novel drug-radiotherapy combinations: recommendations from NRG Oncology, the American Society for Radiation Oncology (ASTRO), the American College of Radiology (ACR), the Sarah Cannon Research Institute, and the American College of Radiation Oncology (ACRO). Lancet Oncol 2024; 25:e489-e500. [PMID: 39362260 DOI: 10.1016/s1470-2045(24)00264-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 10/05/2024]
Abstract
NRG Oncology's Developmental Therapeutics and Radiation Therapy Subcommittee assembled an interdisciplinary group of investigators to address barriers to successful early phase clinical trials of novel combination therapies involving radiation. This Policy Review elucidates some of the many challenges associated with study design for early phase trials combining radiotherapy with novel systemic agents, which are distinct from drug-drug combination development and are often overlooked. We also advocate for potential solutions that could mitigate or eliminate some of these barriers, providing examples of specific clinical trial designs that could help facilitate efficient and effective evaluation of novel drug-radiotherapy combinations.
Collapse
Affiliation(s)
- Zachary S Zumsteg
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Siddharth Sheth
- Division of Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Krishnan R Patel
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Randall J Kimple
- Department of Human Oncology, Univeristy of Wisconsin, Madison, WI, USA
| | | | - Meng Xu-Welliver
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Pedro A Torres-Saavedra
- Division of Cancer Treatment and Diagnosis, Biometric Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Arta M Monjazeb
- Department of Radiation Oncology, University of California, San Diego, CA, USA
| | - Jyoti Mayadev
- Department of Radiation Medicine and Applied Sciences, University of California, San Diego, CA, USA
| | - Steven E Finkelstein
- The US Oncology Network, Florida Cancer Affiliates, Panama City, FL, USA; Sarah Cannon Research Institute, Nashville, TN, USA; Associated Medical Professional of NY, US Urology Partners, Syracuse, NY, USA
| | - John M Buatti
- Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
| | - Sandip P Patel
- Division of Medical Oncology, University of California, San Diego, CA, USA
| | - Steven H Lin
- Department of Thoracic Radiation Oncology, Division of Radiation Oncology, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
5
|
Harary PM, Hori YS, Persad ARL, Tayag A, Ustrzynski L, Emrich SC, Rahimy E, Park DJ, Li G, Chang SD. KEAP1-mutant atypical meningioma: illustrative case. JOURNAL OF NEUROSURGERY. CASE LESSONS 2024; 8:CASE24387. [PMID: 39250830 PMCID: PMC11404106 DOI: 10.3171/case24387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 07/19/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND While genetic testing of tumors is commonly used to inform the selection of systemic therapies, there is limited evidence for the application of radiotherapy for brain cancer. Recent studies have shown that Kelch-like ECH-associated protein 1 (KEAP1), a key regulator of cellular responses to oxidative and electrophilic stress, is associated with radioresistance in multiple cancer types. Several studies have reported the clinical significance of KEAP1 mutation in brain metastasis; however, the effect of KEAP1 mutations on radioresponse in meningioma has never been reported. OBSERVATIONS The authors present the case of a 40-year-old female with a KEAP1 mutation-positive atypical meningioma that was initially treated with resection followed by intensity-modulated radiation therapy (IMRT). Recurrence was observed at 15 months, requiring reoperation and adjuvant stereotactic radiosurgery (SRS). An excellent treatment response was observed at 7 months post-SRS with an improvement in reported symptoms, although bevacizumab was required for the resolution of radiation necrosis observed 2 months post-SRS. LESSONS To the authors' knowledge, this is the first report of KEAP1-mutant meningioma, including its clinical course after comprehensive management. Notably, treatment included multimodal radiotherapy with IMRT followed by SRS. SRS led to an excellent treatment response at the 7-month follow-up. However, radiation necrosis developed after both radiotherapy treatments, suggesting that radiological modification can be beneficial in patients with KEAP1 mutations. https://thejns.org/doi/10.3171/CASE24387.
Collapse
Affiliation(s)
- Paul M Harary
- Departments of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Yusuke S Hori
- Departments of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Amit R L Persad
- Departments of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Armine Tayag
- Departments of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Louisa Ustrzynski
- Departments of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Sara C Emrich
- Departments of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Elham Rahimy
- Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - David J Park
- Departments of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Gordon Li
- Departments of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Steven D Chang
- Departments of Neurosurgery, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
6
|
Aktar T, Modak S, Majumder D, Maiti D. A detailed insight into macrophages' role in shaping lung carcinogenesis. Life Sci 2024; 352:122896. [PMID: 38972632 DOI: 10.1016/j.lfs.2024.122896] [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: 03/05/2024] [Revised: 06/24/2024] [Accepted: 07/03/2024] [Indexed: 07/09/2024]
Abstract
Despite significant advancements in cancer treatment in recent decades, the high mortality rate associated with lung cancer remains a significant concern. The development and proper execution of new targeted therapies needs more deep knowledge regarding the lung cancer associated tumour microenvironment. One of the key component of that tumour microenvironment is the lung resident macrophages. Although in normal physiological condition the lung resident macrophages are believed to maintain lung homeostasis, but they may also initiate a vicious inflammatory response in abnormal conditions which is linked to lung cancer development. Depending on the activation pathway, the lung resident macrophages are either of M1 or M2 sub-type. The M1 and M2 sub-types differ significantly in various prospectuses, from phenotypic markers to metabolic pathways. In addition to this generalized classification, the recent advancement of the multiomics technology is able to identify some other sub-types of lung resident macrophages. Researchers have also observed that these different sub-types can manipulate the pathogenesis of lung carcinogenesis in a context dependent manner and can either promote or inhibit the development of lung carcinogenesis upon receiving proper activation. As proper knowledge about the role played by the lung resident macrophages' in shaping the lung carcinogenesis is limited, so the main purpose of this review is to bring all the available information under the same roof. We also elaborated the different mechanisms involved in maintenance of the plasticity of M1/M2 sub-type, as this plasticity can be a good target for lung cancer treatment.
Collapse
Affiliation(s)
- Tamanna Aktar
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India
| | - Snehashish Modak
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India
| | - Debabrata Majumder
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India; Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Debasish Maiti
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India.
| |
Collapse
|
7
|
Nishitsuji K, Mito R, Ikezaki M, Yano H, Fujiwara Y, Matsubara E, Nishikawa T, Ihara Y, Uchimura K, Iwahashi N, Sakagami T, Suzuki M, Komohara Y. Impacts of cytoplasmic p53 aggregates on the prognosis and the transcriptome in lung squamous cell carcinoma. Cancer Sci 2024; 115:2947-2960. [PMID: 39031627 PMCID: PMC11462941 DOI: 10.1111/cas.16252] [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: 10/03/2023] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 07/22/2024] Open
Abstract
The tumor suppressor TP53 gene, the most frequently mutated gene in human cancers, produces the product tumor protein p53, which plays an essential role in DNA damage. p53 protein mutations may contribute to tumorigenesis by loss of tumor suppressive functions and malignancy of cancer cells via gain-of-oncogenic functions. We previously reported that mutant p53 proteins form aggregates and that cytoplasmic p53 aggregates were associated with poor prognosis in human ovarian cancer. However, the prognostic impact of p53 aggregation in other tumors including lung squamous cell carcinoma (SCC) is poorly understood. Here, we demonstrated that lung SCC cases with cytoplasmic p53 aggregates had a significantly poor clinical prognosis. Analysis via patient-derived tumor organoids (PDOs) established from lung SCC patients and possessing cytoplasmic p53 aggregates showed that eliminating cytoplasmic p53 aggregates suppressed cell proliferation. RNA sequencing and transcriptome analysis of p53 aggregate-harboring PDOs indicated multiple candidate pathways involved in p53 aggregate oncogenic functions. With lung SCC-derived cell lines, we found that cytoplasmic p53 aggregates contributed to cisplatin resistance. This study thus shows that p53 aggregates are a predictor of poor prognosis in lung SCC and suggests that detecting p53 aggregates via p53 conventional immunohistochemical analysis may aid patient selection for platinum-based therapy.
Collapse
Affiliation(s)
- Kazuchika Nishitsuji
- Department of Biochemistry, School of MedicineWakayama Medical UniversityWakayamaJapan
- Unité de Glycobiologie Structurale et FonctionnelleUMR 8576 CNRS, Université de LilleVilleneuve d'AscqFrance
| | - Remi Mito
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Department of Respiratory Medicine, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Midori Ikezaki
- Department of Biochemistry, School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Hiromu Yano
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Yukio Fujiwara
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Eri Matsubara
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Department of Thoracic and Breast Surgery, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Taro Nishikawa
- Department of Biochemistry, School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Yoshito Ihara
- Department of Biochemistry, School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Kenji Uchimura
- Unité de Glycobiologie Structurale et FonctionnelleUMR 8576 CNRS, Université de LilleVilleneuve d'AscqFrance
| | - Naoyuki Iwahashi
- Department of Obstetrics and Gynecology, School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Takuro Sakagami
- Department of Respiratory Medicine, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Makoto Suzuki
- Department of Thoracic and Breast Surgery, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
- Center for Metabolic Regulation of Healthy AgingKumamoto UniversityKumamotoJapan
| |
Collapse
|
8
|
Nishijima A, Oda K, Hasegawa K, Koso T, Asada K, Ikeda Y, Taguchi A, Maeda D, Nagae G, Tsuji S, Tatsuno K, Uehara Y, Kurosaki A, Sato S, Tanikawa M, Sone K, Mori M, Ikemura M, Fujiwara K, Ushiku T, Osuga Y, Aburatani H. Integrated genomic/epigenomic analysis stratifies subtypes of clear cell ovarian carcinoma, highlighting their cellular origin. Sci Rep 2024; 14:18797. [PMID: 39138354 PMCID: PMC11322660 DOI: 10.1038/s41598-024-69796-4] [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: 08/12/2023] [Accepted: 08/08/2024] [Indexed: 08/15/2024] Open
Abstract
The cellular origin of clear cell ovarian carcinoma (CCOC), a major histological subtype of ovarian carcinoma remains elusive. Here, we explored the candidate cellular origin and identify molecular subtypes using integrated genomic/epigenomic analysis. We performed whole exome-sequencing, microarray, and DNA methylation array in 78 CCOC samples according to the original diagnosis. The findings revealed that ARID1A and/or PIK3CA mutations were mutually exclusive with DNA repair related genes, including TP53, BRCA1, and ATM. Clustering of CCOC and other ovarian carcinomas (n = 270) with normal tissues from the fallopian tube, ovarian surface epithelium, endometrial epithelium, and pelvic peritoneum mesothelium (PPM) in a methylation array showed that major CCOC subtypes (with ARID1A and/or PIK3CA mutations) were associated with the PPM-lile cluster (n = 64). This cluster was sub-divided into three clusters: (1) mismatch repair (MMR) deficient with tumor mutational burden-high (n = 2), (2) alteration of ARID1A (n = 51), and (3) ARID1A wild-type (n = 11). The remaining samples (n = 14) were subdivided into (4) ovarian surface epithelium-like (n = 11) and (5) fallopian tube-like (considered as high-grade serous histotype; n = 3). Among these, subtypes (1-3) and others (4 and 5) were found to be associated with immunoreactive signatures and epithelial-mesenchymal transition, respectively. These results contribute to the stratification of CCOC into biological subtypes.
Collapse
Affiliation(s)
- Akira Nishijima
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Katsutoshi Oda
- Division of Integrative Genomics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Takahiro Koso
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Kayo Asada
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Yuji Ikeda
- Department of Obstetrics and Gynecology, Nihon University, Tokyo, Japan
| | - Ayumi Taguchi
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Daichi Maeda
- Department of Molecular and Cellular Pathology, Kanazawa University, Ishikawa, Japan
| | - Genta Nagae
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Shingo Tsuji
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Kenji Tatsuno
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Yuriko Uehara
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Akira Kurosaki
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Sho Sato
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Michihiro Tanikawa
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Kenbun Sone
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Mayuyo Mori
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Masako Ikemura
- Department of Pathology, The University of Tokyo, Tokyo, Japan
| | - Keiichi Fujiwara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Tetsuo Ushiku
- Department of Pathology, The University of Tokyo, Tokyo, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Aburatani
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
9
|
Zhao B, Li B, Guo H, Zhao Q, Zhang X, Zhao H, Xue W, Li W, Duan G, Xu S. The correlation between KRAS and TP53 gene mutations and early growth of pulmonary nodules. J Cardiothorac Surg 2024; 19:376. [PMID: 38926874 PMCID: PMC11200870 DOI: 10.1186/s13019-024-02927-0] [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: 02/21/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
PURPOSE The purpose of this study is to investigate whether gene mutations can lead to the growth of malignant pulmonary nodules. METHODS Retrospective analysis was conducted on patients with pulmonary nodules at Hebei Provincial People's Hospital, collecting basic clinical information such as gender, age, BMI, and hematological indicators. According to the inclusion and exclusion criteria, 85 patients with malignant pulmonary nodules were selected for screening, and gene mutation testing was performed on all patient tissues to explore the relationship between gene mutations and the growth of malignant pulmonary nodules. RESULTS There is a correlation between KRAS and TP53 gene mutations and the growth of pulmonary nodules (P < 0.05), while there is a correlation between KRAS and TP53 gene mutations and the growth of pulmonary nodules in the subgroup of invasive malignant pulmonary nodules (P < 0.05). CONCLUSION Mutations in the TP53 gene can lead to the growth of malignant pulmonary nodules and are correlated with the degree of invasion of malignant pulmonary nodules.
Collapse
Affiliation(s)
- Bin Zhao
- Department of Thoracic Surgery, Hebei General Hospital, Shijiazhuang, 050057, People's Republic of China
- Graduate School, Hebei Medical University, Shijiazhuang, 050011, People's Republic of China
| | - Bin Li
- Hebei Bio-High Technology Development CO., LTD, Shijiazhuang, 050011, People's Republic of China
| | - Haoxin Guo
- Graduate School, Hebei Medical University, Shijiazhuang, 050011, People's Republic of China
| | - Qingtao Zhao
- Department of Thoracic Surgery, Hebei General Hospital, Shijiazhuang, 050057, People's Republic of China
| | - Xiaopeng Zhang
- Department of Thoracic Surgery, Hebei General Hospital, Shijiazhuang, 050057, People's Republic of China
| | - Huanfen Zhao
- Department of Thoracic Surgery, Hebei General Hospital, Shijiazhuang, 050057, People's Republic of China
| | - Wenfei Xue
- Department of Thoracic Surgery, Hebei General Hospital, Shijiazhuang, 050057, People's Republic of China
| | - Wei Li
- Department of Thoracic Surgery, Hebei General Hospital, Shijiazhuang, 050057, People's Republic of China
| | - Guochen Duan
- Department of Thoracic Surgery, Children's Hospital of Hebei Province, Shijiazhuang, 050000, People's Republic of China.
| | - Shun Xu
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, 110000, People's Republic of China
| |
Collapse
|
10
|
Kuchur O, Pogodaeva S, Shcherbakova A, Tsymbal S. Atox1-cyclin D1 loop activity is critical for survival of tumor cells with inactivated TP53. Biosci Rep 2024; 44:BSR20240389. [PMID: 38813981 PMCID: PMC11166628 DOI: 10.1042/bsr20240389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 05/31/2024] Open
Abstract
The search for relevant molecular targets is one of the main tasks of modern tumor chemotherapy. To successfully achieve this, it is necessary to have the most complete understanding of the functioning of a transcriptional apparatus of the cell, particularly related to proliferation. The p53 protein plays an important role in regulating processes such as apoptosis, repair, and cell division, and the loss of its functionality often accompanies various types of tumors and contributes to the development of chemoresistance. Additionally, the proliferative activity of tumor cells is closely related to the metabolism of transition metals. For example, the metallochaperone Atox1 - a copper transporter protein - acts as a transcription activator for cyclin D1, promoting progression through the G1/S phase of the cell cycle. On the other hand, p53 suppresses cyclin D1 at the transcriptional level, thereby these proteins have divergent effects on cell cycle progression. However, the contribution of the interaction between these proteins to cell survival is poorly understood. This work demonstrates that not only exists a positive feedback loop between Atox1 and cyclin D1 but also that the activity of this loop depends on the status of the TP53 gene. Upon inactivation of TP53 in A549 and HepG2 cell lines, the expression of ATOX1 and CCND1 genes is enhanced, and their suppression in these cells leads to pronounced apoptosis. This fundamental observation may be useful in selecting more precise interventions for combined therapy of p53-negative tumors.
Collapse
Affiliation(s)
- Oleg A. Kuchur
- National Research University ITMO, 197101 St. Petersburg, Russia
| | | | | | | |
Collapse
|
11
|
Gilmer TM, Lai CH, Guo K, Deland K, Ashcraft KA, Stewart AE, Wang Y, Fu J, Wood KC, Kirsch DG, Kastan MB. A Novel Dual ATM/DNA-PK Inhibitor, XRD-0394, Potently Radiosensitizes and Potentiates PARP and Topoisomerase I Inhibitors. Mol Cancer Ther 2024; 23:751-765. [PMID: 38588408 DOI: 10.1158/1535-7163.mct-23-0890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/27/2024] [Accepted: 03/25/2024] [Indexed: 04/10/2024]
Abstract
A majority of patients with cancer receive radiotherapy as part of their treatment regimens whether using external beam therapy or locally-delivered radioisotopes. While often effective, some tumors are inadequately controlled with radiation and radiotherapy has significant short-term and long-term toxicities for cancer survivors. Insights into molecular mechanisms involved in cellular responses to DNA breaks introduced by radiation or other cancer therapies have been gained in recent years and approaches to manipulate these responses to enhance tumor cell killing or reduce normal tissue toxicity are of great interest. Here, we report the identification and initial characterization of XRD-0394, a potent and specific dual inhibitor of two DNA damage response kinases, ATM and DNA-PKcs. This orally bioavailable molecule demonstrates significantly enhanced tumor cell kill in the setting of therapeutic ionizing irradiation in vitro and in vivo. XRD-0394 also potentiates the effectiveness of topoisomerase I inhibitors in vitro. In addition, in cells lacking BRCA1/2 XRD-0394 shows single-agent activity and synergy in combination with PARP inhibitors. A phase Ia clinical trial (NCT05002140) with XRD-0394 in combination with radiotherapy has completed. These results provide a rationale for future clinical trials with XRD-0394 in combination with radiotherapy, PARP inhibitors, and targeted delivery of topoisomerase I inhibitors.
Collapse
Affiliation(s)
| | - Chun-Hsiang Lai
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Kexiao Guo
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Katherine Deland
- Department of Radiation Oncology, Duke University School of Medicine, Durham, North Carolina
| | - Kathleen A Ashcraft
- Department of Radiation Oncology, Duke University School of Medicine, Durham, North Carolina
| | - Amy E Stewart
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | | | | | - Kris C Wood
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - David G Kirsch
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
- Department of Radiation Oncology, Duke University School of Medicine, Durham, North Carolina
| | - Michael B Kastan
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
12
|
Liu F, Farris MK, Ververs JD, Hughes RT, Munley MT. Histology-driven hypofractionated radiation therapy schemes for early-stage lung adenocarcinoma and squamous cell carcinoma. Radiother Oncol 2024; 195:110257. [PMID: 38548113 PMCID: PMC11098686 DOI: 10.1016/j.radonc.2024.110257] [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: 01/24/2024] [Revised: 03/06/2024] [Accepted: 03/21/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND AND PURPOSE Histology was found to be an important prognostic factor for local tumor control probability (TCP) after stereotactic body radiotherapy (SBRT) of early-stage non-small-cell lung cancer (NSCLC). A histology-driven SBRT approach has not been explored in routine clinical practice and histology-dependent fractionation schemes remain unknown. Here, we analyzed pooled histologic TCP data as a function of biologically effective dose (BED) to determine histology-driven fractionation schemes for SBRT and hypofractionated radiotherapy of two predominant early-stage NSCLC histologic subtypes adenocarcinoma (ADC) and squamous cell carcinoma (SCC). MATERIAL AND METHODS The least-χ2 method was used to fit the collected histologic TCP data of 8510 early-stage NSCLC patients to determine parameters for a well-developed radiobiological model per the Hypofractionated Treatment Effects in the Clinic (HyTEC) initiative. RESULTS A fit to the histologic TCP data yielded independent radiobiological parameter sets for radiotherapy of early-stage lung ADC and SCC. TCP increases steeply with BED and reaches an asymptotic maximal plateau, allowing us to determine model-independent optimal fractionation schemes of least doses in 1-30 fractions to achieve maximal tumor control for early-stage lung ADC and SCC, e.g., 30, 44, 48, and 51 Gy for ADC, and 32, 48, 54, and 58 Gy for SCC in 1, 3, 4, and 5 fractions, respectively. CONCLUSION We presented the first determination of histology-dependent radiobiological parameters and model-independent histology-driven optimal SBRT and hypofractionated radiation therapy schemes for early-stage lung ADC and SCC. SCC requires substantially higher radiation doses to maximize tumor control than ADC, plausibly attributed to tumor genetic diversity and microenvironment. The determined optimal SBRT schemes agree well with clinical practice for early-stage lung ADC. These proposed optimal fractionation schemes provide first insights for histology-based personalized radiotherapy of two predominant early-stage NSCLC subtypes ADC and SCC, which require further validation with large-scale histologic TCP data.
Collapse
Affiliation(s)
- Feng Liu
- Department of Radiation Oncology, Wake Forest University School of Medicine and Atrium Health Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA.
| | - Michael K Farris
- Department of Radiation Oncology, Wake Forest University School of Medicine and Atrium Health Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA
| | - James D Ververs
- Department of Radiation Oncology, Wake Forest University School of Medicine and Atrium Health Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA
| | - Ryan T Hughes
- Department of Radiation Oncology, Wake Forest University School of Medicine and Atrium Health Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA
| | - Michael T Munley
- Department of Radiation Oncology, Wake Forest University School of Medicine and Atrium Health Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA
| |
Collapse
|
13
|
Ma Z, Men Y, Liu Y, Bao Y, Liu Q, Yang X, Wang J, Deng L, Zhai Y, Bi N, Wang L, Hui Z. Preoperative CT-based radiomic prognostic index to predict the benefit of postoperative radiotherapy in patients with non-small cell lung cancer: a multicenter study. Cancer Imaging 2024; 24:61. [PMID: 38741207 PMCID: PMC11089675 DOI: 10.1186/s40644-024-00707-6] [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: 03/16/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND The value of postoperative radiotherapy (PORT) for patients with non-small cell lung cancer (NSCLC) remains controversial. A subset of patients may benefit from PORT. We aimed to identify patients with NSCLC who could benefit from PORT. METHODS Patients from cohorts 1 and 2 with pathological Tany N2 M0 NSCLC were included, as well as patients with non-metastatic NSCLC from cohorts 3 to 6. The radiomic prognostic index (RPI) was developed using radiomic texture features extracted from the primary lung nodule in preoperative chest CT scans in cohort 1 and validated in other cohorts. We employed a least absolute shrinkage and selection operator-Cox regularisation model for data dimension reduction, feature selection, and the construction of the RPI. We created a lymph-radiomic prognostic index (LRPI) by combining RPI and positive lymph node number (PLN). We compared the outcomes of patients who received PORT against those who did not in the subgroups determined by the LRPI. RESULTS In total, 228, 1003, 144, 422, 19, and 21 patients were eligible in cohorts 1-6. RPI predicted overall survival (OS) in all six cohorts: cohort 1 (HR = 2.31, 95% CI: 1.18-4.52), cohort 2 (HR = 1.64, 95% CI: 1.26-2.14), cohort 3 (HR = 2.53, 95% CI: 1.45-4.3), cohort 4 (HR = 1.24, 95% CI: 1.01-1.52), cohort 5 (HR = 2.56, 95% CI: 0.73-9.02), cohort 6 (HR = 2.30, 95% CI: 0.53-10.03). LRPI predicted OS (C-index: 0.68, 95% CI: 0.60-0.75) better than the pT stage (C-index: 0.57, 95% CI: 0.50-0.63), pT + PLN (C-index: 0.58, 95% CI: 0.46-0.70), and RPI (C-index: 0.65, 95% CI: 0.54-0.75). The LRPI was used to categorize individuals into three risk groups; patients in the moderate-risk group benefited from PORT (HR = 0.60, 95% CI: 0.40-0.91; p = 0.02), while patients in the low-risk and high-risk groups did not. CONCLUSIONS We developed preoperative CT-based radiomic and lymph-radiomic prognostic indexes capable of predicting OS and the benefits of PORT for patients with NSCLC.
Collapse
Affiliation(s)
- Zeliang Ma
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital/National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Men
- Department of VIP Medical Services, National Clinical Research Center for Cancer/Cancer Hospital/National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yunsong Liu
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital/National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongxing Bao
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital/National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qian Liu
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital/National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xu Yang
- Department of Medical Oncology, National Clinical Research Center for Cancer/Cancer Hospital/National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianyang Wang
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital/National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Deng
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital/National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yirui Zhai
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital/National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan Bi
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital/National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Luhua Wang
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital/National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhouguang Hui
- Department of VIP Medical Services, National Clinical Research Center for Cancer/Cancer Hospital/National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
14
|
Cao Y, Fu A, Liu C. Exploring the NRF2-TP53 Signaling Network Through Machine Learning and Pan-Cancer Analysis: Identifying Potential targets for Cancer Prognosis Related to Oxidative Stress. Adv Biol (Weinh) 2024; 8:e2300659. [PMID: 38519438 DOI: 10.1002/adbi.202300659] [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: 12/03/2023] [Revised: 01/24/2024] [Indexed: 03/24/2024]
Abstract
Oxidative stress (OXS) is closely related to tumor prognosis and immune response, while TP53 integrated with NRF2 is closely associated with the regulation of cancer-related OXS. Hence, constructing a TP53-NRF2 integrated OXS signature of pan-cancer is essential in predicting survival prognosis and facilitating cancer drug treatment. The pan-cancer analysis acquired the Cancer Genome Atlas (TCGA) transcriptome sequencing data from UCSC Xena, which consisted of 33 cancer types (n = 10 440). The Random Forest, Lasso regression, and Cox regression analyses are used to construct an OXS score based on 25 OXS genes. Following this, based on the OXS signature, patients are categorized into low- and high-risk groups. The disparities between the two cohorts regarding survival prognosis, immune infiltration, and drug sensitivity are delved deeply. The expression level of genes is confirmed using immunohistochemistry. The prognosis of pan-cancer patients is adequately predicted by the OXS signature with the assistance of the machine-learning algorithm. A highly accurate nomogram is developed by combining the OXS signature and clinical features. The presence of immune cells indicated that the OXS signature can be associated with the critical pathways of immunotherapy for all types of cancer, and BCL2 showed promising results. Distinct inter-group differences are observed in the OXS signature for frequently utilized antineoplastic medications in clinical settings, including first-line drugs suggested in the guidelines. In summary, by conducting a thorough analysis of OXS genes, a new model based on OXSscore is successfully developed. This model can predict the clinical prognosis and drug sensitivity of pan-cancer with high accuracy. Potential stars in the field of cancer-related anti-OXS may include drugs that target BCL2.
Collapse
Affiliation(s)
- Yuchen Cao
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China
| | - Ao Fu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China
| | - Chunjun Liu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China
| |
Collapse
|
15
|
Takahashi J, Suzuki T, Sato M, Nitta S, Yaguchi N, Muta T, Tsuchida K, Suda H, Morita M, Hamada S, Masamune A, Takahashi S, Kamei T, Yamamoto M. Differential squamous cell fates elicited by NRF2 gain of function versus KEAP1 loss of function. Cell Rep 2024; 43:114104. [PMID: 38602872 DOI: 10.1016/j.celrep.2024.114104] [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: 10/16/2023] [Revised: 02/19/2024] [Accepted: 03/27/2024] [Indexed: 04/13/2024] Open
Abstract
Clinical evidence has revealed that high-level activation of NRF2 caused by somatic mutations in NRF2 (NFE2L2) is frequently detected in esophageal squamous cell carcinoma (ESCC), whereas that caused by somatic mutations in KEAP1, a negative regulator of NRF2, is not. Here, we aspire to generate a mouse model of NRF2-activated ESCC using the cancer-derived NRF2L30F mutation and cancer driver mutant TRP53R172H. Concomitant expression of NRF2L30F and TRP53R172H results in formation of NRF2-activated ESCC-like lesions. In contrast, while squamous-cell-specific deletion of KEAP1 induces similar NRF2 hyperactivation, the loss of KEAP1 combined with expression of TRP53R172H does not elicit the formation of ESCC-like lesions. Instead, KEAP1-deleted cells disappear from the esophageal epithelium over time. These findings demonstrate that, while cellular NRF2 levels are similarly induced, NRF2 gain of function and KEAP1 loss of function elicits distinct fates of squamous cells. The NRF2L30F mutant mouse model developed here will be instrumental in elucidating the mechanistic basis leading to NRF2-activated ESCC.
Collapse
Affiliation(s)
- Jun Takahashi
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Japan; Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takafumi Suzuki
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Japan.
| | - Miu Sato
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Japan
| | - Shuji Nitta
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Japan
| | - Nahoko Yaguchi
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Japan
| | - Tatsuki Muta
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Japan
| | - Kouhei Tsuchida
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Japan
| | - Hiromi Suda
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Japan
| | - Masanobu Morita
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Japan
| | - Shin Hamada
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center in Transborder Medical Research Center, University of Tsukuba, Tsukuba, Japan
| | - Takashi Kamei
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masayuki Yamamoto
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Japan.
| |
Collapse
|
16
|
Li Q, Song Q, Pei H, Chen Y. Emerging mechanisms of ferroptosis and its implications in lung cancer. Chin Med J (Engl) 2024; 137:818-829. [PMID: 38494343 PMCID: PMC10997236 DOI: 10.1097/cm9.0000000000003048] [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: 06/28/2023] [Indexed: 03/19/2024] Open
Abstract
ABSTRACT Lung cancer is one of the most common malignancies and has the highest number of deaths among all cancers. Despite continuous advances in medical strategies, the overall survival of lung cancer patients is still low, probably due to disease progression or drug resistance. Ferroptosis is an iron-dependent form of regulated cell death triggered by the lethal accumulation of lipid peroxides, and its dysregulation is implicated in cancer development. Preclinical evidence has shown that targeting the ferroptosis pathway could be a potential strategy for improving lung cancer treatment outcomes. In this review, we summarize the underlying mechanisms and regulatory networks of ferroptosis in lung cancer and highlight ferroptosis-targeting preclinical attempts to provide new insights for lung cancer treatment.
Collapse
Affiliation(s)
- Qian Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Qibin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Huadong Pei
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington D.C. 20057, USA
| | - Yali Chen
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| |
Collapse
|
17
|
Sitthideatphaiboon P, Nantavithya C, Chantranuwat P, Vinayanuwattikun C, Sriuranpong V. Impact of LKB1 status on radiation outcome in patients with stage III non-small-cell lung cancer. Sci Rep 2024; 14:6146. [PMID: 38480816 PMCID: PMC10938003 DOI: 10.1038/s41598-024-55476-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/23/2024] [Indexed: 03/17/2024] Open
Abstract
Preclinical studies suggest that loss of LKB1 expression renders cancer cells less responsive to radiation partly through NRF2-mediated upregulation of antioxidant enzymes protecting against radiation-induced DNA damage. Here we investigated the association of an alteration in this pathway with radio-resistance in lung cancer patients. Patients with locally advanced non-small cell lung cancer (LA-NSCLC) who were treated with chemoradiotherapy (CRT) and analyzed for LKB1 expression using semiquantitative immunohistochemistry. Clinical characteristics and expression of LKB1 were analyzed for association with radiotherapy outcomes. We analyzed 74 available tumor specimens from 178 patients. After a median follow-up of 40.7 months, 2-year cumulative incidence of locoregional recurrence (LRR) in patients who had LKB1Low expression was significantly higher than those with LKB1High expression (68.8% vs. 31.3%, P = 0.0001). LKB1Low expression was found significantly associated with a higher incidence of distant metastases (DM) (P = 0.0008), shorter disease-free survival (DFS) (P = 0.006), and worse overall survival (OS) (P = 0.02) compared to LKB1High expression. Moreover, patients with LKB1Low expression showed a significantly higher 2-year cumulative incidence of LRR (77.6% vs. 21%; P = 0.02), higher DM recurrence (P = 0.002), and shorter OS (P < 0.0001) compared with the EGFR-mutant group. For all patients with LKB1Low who had LRR, these recurrences occurred within the field of radiation, in contrast to those with LKB1High expression having both in-field, marginal, and out-of-field failures. LKB1 expression may serve as a potential biomarker for poor outcomes after receiving radiation in LA-NSCLC patients. Further studies to confirm the association and application are warranted.
Collapse
Affiliation(s)
- Piyada Sitthideatphaiboon
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and the King Chulalongkorn Memorial Hospital, 1873 Henry Dunant Rd, Pathumwan, Bangkok, 10330, Thailand
| | - Chonnipa Nantavithya
- Division of Therapeutic Radiation and Oncology, Department of Radiology, Faculty of Medicine, Chulalongkorn University and the King Chulalongkorn Memorial Hospital, Bangkok, 10330, Thailand
| | - Poonchavist Chantranuwat
- Department of Pathology, Faculty of Medicine, Chulalongkorn University and the King Chulalongkorn Memorial Hospital, Bangkok, 10330, Thailand
| | - Chanida Vinayanuwattikun
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and the King Chulalongkorn Memorial Hospital, 1873 Henry Dunant Rd, Pathumwan, Bangkok, 10330, Thailand
| | - Virote Sriuranpong
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and the King Chulalongkorn Memorial Hospital, 1873 Henry Dunant Rd, Pathumwan, Bangkok, 10330, Thailand.
| |
Collapse
|
18
|
Bae T, Hallis SP, Kwak MK. Hypoxia, oxidative stress, and the interplay of HIFs and NRF2 signaling in cancer. Exp Mol Med 2024; 56:501-514. [PMID: 38424190 PMCID: PMC10985007 DOI: 10.1038/s12276-024-01180-8] [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: 11/14/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 03/02/2024] Open
Abstract
Oxygen is crucial for life and acts as the final electron acceptor in mitochondrial energy production. Cells adapt to varying oxygen levels through intricate response systems. Hypoxia-inducible factors (HIFs), including HIF-1α and HIF-2α, orchestrate the cellular hypoxic response, activating genes to increase the oxygen supply and reduce expenditure. Under conditions of excess oxygen and resulting oxidative stress, nuclear factor erythroid 2-related factor 2 (NRF2) activates hundreds of genes for oxidant removal and adaptive cell survival. Hypoxia and oxidative stress are core hallmarks of solid tumors and activated HIFs and NRF2 play pivotal roles in tumor growth and progression. The complex interplay between hypoxia and oxidative stress within the tumor microenvironment adds another layer of intricacy to the HIF and NRF2 signaling systems. This review aimed to elucidate the dynamic changes and functions of the HIF and NRF2 signaling pathways in response to conditions of hypoxia and oxidative stress, emphasizing their implications within the tumor milieu. Additionally, this review explored the elaborate interplay between HIFs and NRF2, providing insights into the significance of these interactions for the development of novel cancer treatment strategies.
Collapse
Affiliation(s)
- Taegeun Bae
- Integrated Research Institute for Pharmaceutical Sciences, The Catholic University of Korea, Bucheon, Gyeonggi‑do, 14662, Republic of Korea
| | - Steffanus Pranoto Hallis
- Department of Pharmacy, Graduate School of The Catholic University of Korea, Bucheon, Gyeonggi‑do, 14662, Republic of Korea
| | - Mi-Kyoung Kwak
- Integrated Research Institute for Pharmaceutical Sciences, The Catholic University of Korea, Bucheon, Gyeonggi‑do, 14662, Republic of Korea.
- Department of Pharmacy, Graduate School of The Catholic University of Korea, Bucheon, Gyeonggi‑do, 14662, Republic of Korea.
- College of Pharmacy, The Catholic University of Korea, Bucheon, Gyeonggi‑do, 14662, Republic of Korea.
| |
Collapse
|
19
|
Bourbonne V, Morjani M, Pradier O, Hatt M, Jaouen V, Querellou S, Visvikis D, Lucia F, Schick U. PET/CT-Based Radiogenomics Supports KEAP1/NFE2L2 Pathway Targeting for Non-Small Cell Lung Cancer Treated with Curative Radiotherapy. J Nucl Med 2024:jnumed.123.266749. [PMID: 38360055 DOI: 10.2967/jnumed.123.266749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/02/2024] [Indexed: 02/17/2024] Open
Abstract
In lung cancer patients, radiotherapy is associated with a increased risk of local relapse (LR) when compared with surgery but with a preferable toxicity profile. The KEAP1/NFE2L2 mutational status (MutKEAP1/NFE2L2) is significantly correlated with LR in patients treated with radiotherapy but is rarely available. Prediction of MutKEAP1/NFE2L2 with noninvasive modalities could help to further personalize each therapeutic strategy. Methods: Based on a public cohort of 770 patients, model RNA (M-RNA) was first developed using continuous gene expression levels to predict MutKEAP1/NFE2L2, resulting in a binary output. The model PET/CT (M-PET/CT) was then built to predict M-RNA binary output using PET/CT-extracted radiomics features. M-PET/CT was validated on an external cohort of 151 patients treated with curative volumetric modulated arc radiotherapy. Each model was built, internally validated, and evaluated on a separate cohort using a multilayer perceptron network approach. Results: The M-RNA resulted in a C statistic of 0.82 in the testing cohort. With a training cohort of 101 patients, the retained M-PET/CT resulted in an area under the curve of 0.90 (P < 0.001). With a probability threshold of 20% applied to the testing cohort, M-PET/CT achieved a C statistic of 0.7. The same radiomics model was validated on the volumetric modulated arc radiotherapy cohort as patients were significantly stratified on the basis of their risk of LR with a hazard ratio of 2.61 (P = 0.02). Conclusion: Our approach enables the prediction of MutKEAP1/NFE2L2 using PET/CT-extracted radiomics features and efficiently classifies patients at risk of LR in an external cohort treated with radiotherapy.
Collapse
Affiliation(s)
- Vincent Bourbonne
- Department of Radiation Oncology, University Hospital, Brest, France;
- LaTIM UMR 1101 INSERM, University Brest, Brest, France
| | - Moncef Morjani
- Department of Radiation Oncology, University Hospital, Brest, France
| | - Olivier Pradier
- Department of Radiation Oncology, University Hospital, Brest, France
- LaTIM UMR 1101 INSERM, University Brest, Brest, France
| | - Mathieu Hatt
- LaTIM UMR 1101 INSERM, University Brest, Brest, France
| | - Vincent Jaouen
- LaTIM UMR 1101 INSERM, University Brest, Brest, France
- Institut Mines-Télécom Atlantique, Brest, France
| | - Solène Querellou
- Nuclear Medicine Department, University Hospital, Brest, France; and
- Groupe d'Étude de la Thrombose Occidentale GETBO (INSERM UMR 1304), Université de Bretagne Occidentale, Brest, France
| | | | - François Lucia
- Department of Radiation Oncology, University Hospital, Brest, France
- LaTIM UMR 1101 INSERM, University Brest, Brest, France
| | - Ulrike Schick
- Department of Radiation Oncology, University Hospital, Brest, France
- LaTIM UMR 1101 INSERM, University Brest, Brest, France
| |
Collapse
|
20
|
Chen F, Naughton KJ, Lee JH, Brainson CF. Using 3-Dimensional Cultures to Propagate Genetically Modified Lung Organoids. Methods Mol Biol 2024; 2805:19-30. [PMID: 39008172 DOI: 10.1007/978-1-0716-3854-5_2] [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] [Indexed: 07/16/2024]
Abstract
Transformed lung organoids have extensive applications in lung cancer modeling and drug screening. Traditional two-dimensional (2D) cultures fail to propagate a large subpopulation of murine primary tumors in vitro. However, three-dimensional (3D) air-liquid interface (ALI) cultures, which are employed to grow normal lung organoids, can be used to efficiently culture cancerous lung tumor cells. Here, we detail a procedure for cultivating genetically modified lung organoids in 3D-ALI cultures. This protocol contains two parts. The first part describes how to transduce lung epithelial cells, which are either freshly sorted from lungs or from actively growing murine organoids, with virus in order to modify gene expression. The target lung cells are incubated with virus for 1-2 h for transduction. Then, the transduced cells are thoroughly washed and mixed with stromal support cells and Matrigel and are loaded into transwell inserts for culture and validated for genetic modifications through downstream assays. The second part describes how to isolate tumor cells growing orthotopically in genetically engineered mouse models to produce organoid cell lines that can be used for ex vivo drug discovery assays. For this protocol, tumors are isolated from lungs of mice, finely chopped and washed. Then, tumor chunks are mixed with Matrigel for 3D-ALI culture. Finally, organoids budding from tumor chunks are trypsinized and passaged to establish an organoid line. Together these two protocols provide a promising platform to study the genesis, progression, and treatment of lung cancer.
Collapse
Affiliation(s)
- Fan Chen
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, P. R. China
| | - Kassandra J Naughton
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Joo-Hyeon Lee
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Christine F Brainson
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA.
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
21
|
Moubarak MM, Pagano Zottola AC, Larrieu CM, Cuvellier S, Daubon T, Martin OCB. Exploring the multifaceted role of NRF2 in brain physiology and cancer: A comprehensive review. Neurooncol Adv 2024; 6:vdad160. [PMID: 38221979 PMCID: PMC10785770 DOI: 10.1093/noajnl/vdad160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024] Open
Abstract
Chronic oxidative stress plays a critical role in the development of brain malignancies due to the high rate of brain oxygen utilization and concomitant production of reactive oxygen species. The nuclear factor-erythroid-2-related factor 2 (NRF2), a master regulator of antioxidant signaling, is a key factor in regulating brain physiology and the development of age-related neurodegenerative diseases. Also, NRF2 is known to exert a protective antioxidant effect against the onset of oxidative stress-induced diseases, including cancer, along with its pro-oncogenic activities through regulating various signaling pathways and downstream target genes. In glioblastoma (GB), grade 4 glioma, tumor resistance, and recurrence are caused by the glioblastoma stem cell population constituting a small bulk of the tumor core. The persistence and self-renewal capacity of these cell populations is enhanced by NRF2 expression in GB tissues. This review outlines NRF2's dual involvement in cancer and highlights its regulatory role in human brain physiology and diseases, in addition to the development of primary brain tumors and therapeutic potential, with a focus on GB.
Collapse
Affiliation(s)
- Maya M Moubarak
- University of Bordeaux, CNRS, IBGC, UMR 5095, Bordeaux, France
| | | | | | | | - Thomas Daubon
- University of Bordeaux, CNRS, IBGC, UMR 5095, Bordeaux, France
| | | |
Collapse
|
22
|
Ngo HKC, Le H, Surh YJ. Nrf2, A Target for Precision Oncology in Cancer Prognosis and Treatment. J Cancer Prev 2023; 28:131-142. [PMID: 38205365 PMCID: PMC10774478 DOI: 10.15430/jcp.2023.28.4.131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Activating nuclear factor-erythroid 2-related factor (Nrf2), a master regulator of redox homeostasis, has been shown to suppress initiation of carcinogenesis in normal cells. However, this transcription factor has recently been reported to promote proliferation of some transformed or cancerous cells. In tumor cells, Nrf2 is prone to mutations that result in stabilization and concurrent accumulation of its protein product. A hyperactivated mutant form of Nrf2 could support the cancer cells for enhanced proliferation, invasiveness, and resistance to chemotherapeutic agents and radiotherapy, which are associated with a poor clinical outcome. Hence understanding mutations in Nrf2 would have a significant impact on the prognosis and treatment of cancer in the era of precision medicine. This perspective would provide an insight into the genetic alterations in Nrf2 and suggest the application of small molecules, RNAi, and genome editing technologies, particularly CRISR-Cas9, in therapeutic intervention of cancer in the context of the involvement of Nrf2 mutations.
Collapse
Affiliation(s)
- Hoang Kieu Chi Ngo
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Hoang Le
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Young-Joon Surh
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, Korea
- Cancer Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
23
|
Duan J, Zhang Y, Chen R, Liang L, Huo Y, Lu S, Zhao J, Hu C, Sun Y, Yang K, Chen M, Yu Y, Ying J, Huang R, Ma X, Leaw S, Bai F, Shen Z, Cai S, Gao D, Wang J, Wang Z. Tumor-immune microenvironment and NRF2 associate with clinical efficacy of PD-1 blockade combined with chemotherapy in lung squamous cell carcinoma. Cell Rep Med 2023; 4:101302. [PMID: 38052215 PMCID: PMC10772345 DOI: 10.1016/j.xcrm.2023.101302] [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: 09/30/2022] [Revised: 03/29/2023] [Accepted: 11/02/2023] [Indexed: 12/07/2023]
Abstract
The RATIONALE-307 study (ClinicalTrials.gov: NCT03594747) demonstrates prolonged progression-free survival (PFS) with first-line tislelizumab plus chemotherapy versus chemotherapy in advanced lung squamous cell carcinoma (LUSC; N = 360). Here we describe an immune-related gene expression signature (GES), composed of genes involved in both innate and adaptive immunity, that appears to differentiate tislelizumab plus chemotherapy PFS benefit versus chemotherapy. In contrast, a tislelizumab plus chemotherapy PFS benefit is observed regardless of programmed death ligand 1 (PD-L1) expression or tumor mutational burden (TMB). Genetic analysis reveals that NRF2 pathway activation is enriched in PD-L1positive and TMBhigh patients. NRF2 pathway activation is negatively associated with PFS, which affects efficacy outcomes associated with PD-L1 and TMB status, impairing their predictive potential. Mechanistic studies demonstrate that NRF2 directly mediates PD-L1 constitutive expression independent of adaptive PD-L1 regulation in LUSC. In summary, the GES is an immune signature that might identify LUSC patients likely to benefit from first-line tislelizumab plus chemotherapy.
Collapse
Affiliation(s)
- Jianchun Duan
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yun Zhang
- BeiGene (Beijing) Co., Ltd., Beijing 100022, China
| | - Ran Chen
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, China; Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200090, China
| | - Liang Liang
- BeiGene (Beijing) Co., Ltd., Beijing 100022, China
| | - Yi Huo
- BeiGene (Beijing) Co., Ltd., Beijing 100022, China
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Jun Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Chunhong Hu
- Oncology Department, The Second Hospital of Central South University, Changsha 410011, China
| | - Yuping Sun
- Oncology Department, Jinan Central Hospital, Shandong 250013, China
| | - Kunyu Yang
- Union Hospital, Cancer Center, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430074, China
| | - Mingwei Chen
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yan Yu
- Department of Respiratory Medicine, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ruiqi Huang
- BeiGene (Shanghai) Co., Ltd., Shanghai 200040, China
| | - Xiaopeng Ma
- BeiGene (Beijing) Co., Ltd., Beijing 100022, China
| | | | - Fan Bai
- BeiGene (Shanghai) Co., Ltd., Shanghai 200040, China
| | - Zhirong Shen
- BeiGene (Beijing) Co., Ltd., Beijing 100022, China
| | - Shangli Cai
- Burning Rock Biotech, Guangzhou 510300, China
| | - Daming Gao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; University of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China; School of Life Science, Hangzhou Institute for Advanced Study, University of the Chinese Academy of Sciences, Hangzhou 310024, China
| | - Jie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Zhijie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
24
|
Greenwood HE, Edwards RS, Tyrrell WE, Barber AR, Baark F, Tanc M, Khalil E, Falzone A, Ward NP, DeBlasi JM, Torrente L, Pearce DR, Firth G, Smith LM, Timmermand OV, Huebner A, George ME, Swanton C, Hynds RE, DeNicola GM, Witney TH. Imaging the master regulator of the antioxidant response in non-small cell lung cancer with positron emission tomography. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.16.572007. [PMID: 38168428 PMCID: PMC10760199 DOI: 10.1101/2023.12.16.572007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Mutations in the NRF2-KEAP1 pathway are common in non-small cell lung cancer (NSCLC) and confer broad-spectrum therapeutic resistance, leading to poor outcomes. The cystine/glutamate antiporter, system xc-, is one of the >200 cytoprotective proteins controlled by NRF2, which can be non-invasively imaged by (S)-4-(3-18F-fluoropropyl)-l-glutamate ([18F]FSPG) positron emission tomography (PET). Through genetic and pharmacologic manipulation, we show that [18F]FSPG provides a sensitive and specific marker of NRF2 activation in advanced preclinical models of NSCLC. We validate imaging readouts with metabolomic measurements of system xc- activity and their coupling to intracellular glutathione concentration. A redox gene signature was measured in patients from the TRACERx 421 cohort, suggesting an opportunity for patient stratification prior to imaging. Furthermore, we reveal that system xc- is a metabolic vulnerability that can be therapeutically targeted for sustained tumour growth suppression in aggressive NSCLC. Our results establish [18F]FSPG as predictive marker of therapy resistance in NSCLC and provide the basis for the clinical evaluation of both imaging and therapeutic agents that target this important antioxidant pathway.
Collapse
Affiliation(s)
- Hannah E. Greenwood
- School of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, UK
| | - Richard S. Edwards
- School of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, UK
| | - Will E. Tyrrell
- School of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, UK
| | - Abigail R. Barber
- School of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, UK
| | - Friedrich Baark
- School of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, UK
| | - Muhammet Tanc
- School of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, UK
| | - Eman Khalil
- School of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, UK
| | - Aimee Falzone
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Nathan P. Ward
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Janine M. DeBlasi
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Laura Torrente
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - David R. Pearce
- CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, University College London, WC1E 6DD, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - George Firth
- School of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, UK
| | - Lydia M. Smith
- School of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, UK
| | - Oskar Vilhelmsson Timmermand
- School of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, UK
| | - Ariana Huebner
- CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, University College London, WC1E 6DD, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Madeleine E. George
- School of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, UK
| | - Charles Swanton
- CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, University College London, WC1E 6DD, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Robert E. Hynds
- CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, University College London, WC1E 6DD, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Gina M. DeNicola
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Timothy H. Witney
- School of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, UK
| |
Collapse
|
25
|
Kwon J, Zhang J, Mok B, Allsup S, Kim C, Toretsky J, Han C. USP13 drives lung squamous cell carcinoma by switching lung club cell lineage plasticity. Mol Cancer 2023; 22:204. [PMID: 38093367 PMCID: PMC10717271 DOI: 10.1186/s12943-023-01892-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/27/2023] [Indexed: 12/17/2023] Open
Abstract
Lung squamous cell carcinoma (LUSC) is associated with high mortality and limited targeted therapies. USP13 is one of the most amplified genes in LUSC, yet its role in lung cancer is largely unknown. Here, we established a novel mouse model of LUSC by overexpressing USP13 on KrasG12D/+; Trp53flox/flox background (KPU). KPU-driven lung squamous tumors faithfully recapitulate key pathohistological, molecular features, and cellular pathways of human LUSC. We found that USP13 altered lineage-determining factors such as NKX2-1 and SOX2 in club cells of the airway and reinforced the fate of club cells to squamous carcinoma development. We showed a strong molecular association between USP13 and c-MYC, leading to the upregulation of squamous programs in murine and human lung cancer cells. Collectively, our data demonstrate that USP13 is a molecular driver of lineage plasticity in club cells and provide mechanistic insight that may have potential implications for the treatment of LUSC.
Collapse
Affiliation(s)
- Juntae Kwon
- Department of Oncology, Georgetown University School of Medicine, Washington D.C, USA
| | - Jinmin Zhang
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington D.C, USA
| | - Boram Mok
- Department of Oncology, Georgetown University School of Medicine, Washington D.C, USA
| | - Samuel Allsup
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington D.C, USA
| | - Chul Kim
- Division of Hematology and Oncology, Georgetown University School of Medicine, Washington D.C, USA
- MedStar Georgetown University Hospital, Washington D.C, USA
- Lombardi Comprehensive Cancer Center, Washington D.C, USA
| | - Jeffrey Toretsky
- Department of Oncology, Georgetown University School of Medicine, Washington D.C, USA
- Lombardi Comprehensive Cancer Center, Washington D.C, USA
- Departments of Pediatrics, Washington D.C, USA
| | - Cecil Han
- Department of Oncology, Georgetown University School of Medicine, Washington D.C, USA.
- Lombardi Comprehensive Cancer Center, Washington D.C, USA.
| |
Collapse
|
26
|
Jagasia S, Tasci E, Zhuge Y, Camphausen K, Krauze AV. Identifying patients suitable for targeted adjuvant therapy: advances in the field of developing biomarkers for tumor recurrence following irradiation. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2023; 8:33-42. [PMID: 37982134 PMCID: PMC10655913 DOI: 10.1080/23808993.2023.2276927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/25/2023] [Indexed: 11/21/2023]
Abstract
Introduction Radiation therapy (RT) is commonly used to treat cancer in conjunction with chemotherapy, immunotherapy, and targeted therapies. Despite the effectiveness of RT, tumor recurrence due to treatment resistance still lead to treatment failure. RT-specific biomarkers are currently lacking and remain challenging to investigate with existing data since, for many common malignancies, standard of care (SOC) paradigms involve the administration of RT in conjunction with other agents. Areas Covered Established clinically relevant biomarkers are used in surveillance, as prognostic indicators, and sometimes for treatment planning; however, the inability to intercept early recurrence or predict upfront resistance to treatment remains a significant challenge that limits the selection of patients for adjuvant therapy. We discuss attempts at intercepting early failure. We examine biomarkers that have made it into the clinic where they are used for treatment monitoring and management alteration, and novel biomarkers that lead the field with targeted adjuvant therapy seeking to harness these. Expert Opinion Given the growth of data correlating interventions with omic analysis toward identifying biomarkers of radiation resistance, more robust markers of recurrence that link to biology will increasingly be leveraged toward targeted adjuvant therapy to make a successful transition to the clinic in the coming years.
Collapse
Affiliation(s)
- S Jagasia
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA
| | - E Tasci
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA
| | - Ying Zhuge
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA
| | - K Camphausen
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA
| | - A V Krauze
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Building 10, CRC, Bethesda, MD 20892, USA
| |
Collapse
|
27
|
Gensheimer MF, Gee H, Shirato H, Taguchi H, Snyder JM, Chin AL, Vitzthum LK, Maxim PG, Wakelee HA, Neal J, Das M, Chang DT, Kidd E, Hancock SL, Shultz DB, Horst KC, Le QT, Wong S, Brown E, Nguyen N, Liang R, Loo BW, Diehn M. Individualized Stereotactic Ablative Radiotherapy for Lung Tumors: The iSABR Phase 2 Nonrandomized Controlled Trial. JAMA Oncol 2023; 9:1525-1534. [PMID: 37707820 PMCID: PMC10502697 DOI: 10.1001/jamaoncol.2023.3495] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 06/11/2023] [Indexed: 09/15/2023]
Abstract
Importance Stereotactic ablative radiotherapy (SABR) is used for treating lung tumors but can cause toxic effects, including life-threatening damage to central structures. Retrospective data suggested that small tumors up to 10 cm3 in volume can be well controlled with a biologically effective dose less than 100 Gy. Objective To assess whether individualizing lung SABR dose and fractionation by tumor size, location, and histological characteristics may be associated with local tumor control. Design, Setting, and Participants This nonrandomized controlled trial (the iSABR trial, so named for individualized SABR) was a phase 2 multicenter trial enrolling participants from November 15, 2011, to December 5, 2018, at academic medical centers in the US and Japan. Data were analyzed from December 9, 2020, to May 10, 2023. Patients were enrolled in 3 groups according to cancer type: initial diagnosis of non-small cell lung cancer (NSCLC) with an American Joint Committee on Cancer 7th edition T1-3N0M0 tumor (group 1), a T1-3N0M0 new primary NSCLC with a history of prior NSCLC or multiple NSCLCs (group 2), or lung metastases from NSCLC or another solid tumor (group 3). Intervention Up to 4 tumors were treated with once-daily SABR. The dose ranged from 25 Gy in 1 fraction for peripheral tumors with a volume of 0 to 10 cm3 to 60 Gy in 8 fractions for central tumors with a volume greater than 30 cm3. Main outcome Per-group freedom from local recurrence (same-lobe recurrence) at 1 year, with censoring at time of distant recurrence, death, or loss to follow-up. Results In total, 217 unique patients (median [IQR] age, 72 [64-80] years; 129 [59%] male; 150 [69%] current or former smokers) were enrolled (some multiple times). There were 240 treatment courses: 79 in group 1, 82 in group 2, and 79 in group 3. A total of 285 tumors (211 [74%] peripheral and 74 [26%] central) were treated. The most common dose was 25 Gy in 1 fraction (158 tumors). The median (range) follow-up period was 33 (2-109) months, and the median overall survival was 59 (95% CI, 49-82) months. Freedom from local recurrence at 1 year was 97% (90% CI, 91%-99%) for group 1, 94% (90% CI, 87%-97%) for group 2, and 96% (90% CI, 89%-98%) for group 3. Freedom from local recurrence at 5 years ranged from 83% to 93% in the 3 groups. The proportion of patients with grade 3 to 5 toxic effects was low, at 5% (including a single patient [1%] with grade 5 toxic effects). Conclusions and Relevance The results of this nonrandomized controlled trial suggest that individualized SABR (iSABR) used to treat lung tumors may allow minimization of treatment dose and is associated with excellent local control. Individualized dosing should be considered for use in future trials. Trial Registration ClinicalTrials.gov Identifier: NCT01463423.
Collapse
Affiliation(s)
- Michael F. Gensheimer
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, California
| | - Harriet Gee
- Sydney West Radiation Oncology Network, Sydney, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Hiroki Shirato
- Department of Radiation Oncology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroshi Taguchi
- Department of Radiation Oncology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - John M. Snyder
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, California
| | - Alexander L. Chin
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, California
| | - Lucas K. Vitzthum
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, California
| | - Peter G. Maxim
- Department of Radiation Oncology, University of California Irvine, Irvine, California
| | - Heather A. Wakelee
- Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Joel Neal
- Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Millie Das
- Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Daniel T. Chang
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, California
| | - Elizabeth Kidd
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, California
| | - Steven L. Hancock
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, California
| | - David B. Shultz
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Kathleen C. Horst
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, California
| | - Quynh-Thu Le
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, California
| | - Samantha Wong
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, California
| | - Eleanor Brown
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, California
| | - Ngan Nguyen
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, California
| | - Rachel Liang
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, California
| | - Billy W. Loo
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, California
| | - Maximilian Diehn
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, California
| |
Collapse
|
28
|
Islam SS, Karakas B, Aboussekhra A, Noman ASM. KEAP1/NRF2 Mutations in Stem Cells Define an Aggressive Subset of Head and Neck Cancer Patients Who Have a Poor Prognosis, Lung Metastasis, and Therapeutic Failure. Cancers (Basel) 2023; 15:5006. [PMID: 37894373 PMCID: PMC10605399 DOI: 10.3390/cancers15205006] [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: 08/23/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Mutations in Keap1/Nrf2 in head and neck cancer result in abnormal cell growth. Progenitor cells, bulk tumor cells, and head and neck cancer stem cells (HN-CSCs) may all harbor these mutations. Nevertheless, whether Keap1/Nrf2 mutations in HN-CSCs have an impact on clinical outcomes is unknown. Cancerous HN-CSCs and benign stem cells were obtained from freshly resected head and neck cancer patients (n = 50) via flow cytometry cell sorting and tested for Keap1/Nrf2 mutations. The existence of Keap1/Nrf2 mutations in HN-CSCs, as well as their correlations with tumor mutations, pathologic tumor stage, tumor histologic grades, lung metastasis, treatment outcomes, and the patient's age and conditions, are assessed at the last follow-up visit. Thirteen tumors were found to have Keap1/Nrf2 mutations in their HN-CSCs. More than half of the lung metastases and disease progression occurred in HN-CSCs with mutations. Patients whose tumors carried Keap1/Nrf2 mutations in their HN-CSCs had significantly shorter progression-free survival, overall survival, and time of treatment failure than their non-HN-CSC counterparts. These associations were partly driven by HN-CSCs, in which Keap1/Nrf2 mutations were overrepresented in fast progressors and associated with an increased risk of disease progression. Our findings suggest that molecular genotyping of HN-CSCs may facilitate personalized treatment strategies and assist in identifying patients who are likely to benefit from chemotherapy.
Collapse
Affiliation(s)
- Syed S. Islam
- Department Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
- Faculty of Medicine, Al-Faisal University, Riyadh 11533, Saudi Arabia
| | - Bedri Karakas
- 3 B & B Bio, 4 Professional Drive, Gaithersburg, MD 20879, USA;
| | - Abdelilah Aboussekhra
- Department Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Abu Shadat M. Noman
- Department Biochemistry and Molecular Biology, The University of Chittagong, Chittagong 4331, Bangladesh
| |
Collapse
|
29
|
Xia L, Ma W, Afrashteh A, Sajadi MA, Fakheri H, Valilo M. The nuclear factor erythroid 2-related factor 2/p53 axis in breast cancer. Biochem Med (Zagreb) 2023; 33:030504. [PMID: 37841775 PMCID: PMC10564154 DOI: 10.11613/bm.2023.030504] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023] Open
Abstract
One of the most important factors involved in the response to oxidative stress (OS) is the nuclear factor erythroid 2-related factor 2 (Nrf2), which regulates the expression of components such as antioxidative stress proteins and enzymes. Under normal conditions, Kelch-like ECH-associated protein 1 (Keap1) keeps Nrf2 in the cytoplasm, thus preventing its translocation to the nucleus and inhibiting its role. It has been established that Nrf2 has a dual function; on the one hand, it promotes angiogenesis and cancer cell metastasis while causing resistance to drugs and chemotherapy. On the other hand, Nrf2 increases expression and proliferation of glutathione to protect cells against OS. p53 is a tumour suppressor that activates the apoptosis pathway in aging and cancer cells in addition to stimulating the glutaminolysis and antioxidant pathways. Cancer cells use the antioxidant ability of p53 against OS. Therefore, in the present study, we discussed function of Nrf2 and p53 in breast cancer (BC) cells to elucidate their role in protection or destruction of cancer cells as well as their drug resistance or antioxidant properties.
Collapse
Affiliation(s)
- Lei Xia
- Surgical oncology ward 2, Qinghai Provincial People’s Hospital, Xining Qinghai, China
| | - Wenbiao Ma
- Surgical oncology ward 2, Qinghai Provincial People’s Hospital, Xining Qinghai, China
| | - Ahmad Afrashteh
- Department of Periodontics, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hadi Fakheri
- Paramedical Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Valilo
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
30
|
Lamba N, Cagney DN, Catalano PJ, Kim D, Elhalawani H, Haas-Kogan DA, Wen PY, Wagle N, Aizer AA. A genomic score to predict local control among patients with brain metastases managed with radiation. Neuro Oncol 2023; 25:1815-1827. [PMID: 37260393 PMCID: PMC10547520 DOI: 10.1093/neuonc/noad098] [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: 02/24/2023] [Indexed: 06/02/2023] Open
Abstract
BACKGROUND Clinical predictors of local recurrence following radiation among patients with brain metastases (BrM) provide limited explanatory power. We developed a DNA-based signature of radiotherapeutic efficacy among patients with BrM to better characterize recurrence risk. METHODS We identified 570 patients with 1487 BrM managed with whole-brain (WBRT) or stereotactic radiation therapy at Brigham and Women's Hospital/Dana-Farber Cancer Institute (2013-2020) for whom next-generation sequencing panel data (OncoPanel) were available. Fine/Gray's competing risks regression was utilized to compare local recurrence on a per-metastasis level among patients with versus without somatic alterations of likely biological significance across 84 genes. Genes with a q-value ≤ 0.10 were utilized to develop a "Brain-Radiation Prediction Score" ("Brain-RPS"). RESULTS Genomic alterations in 11 (ATM, MYCL, PALB2, FAS, PRDM1, PAX5, CDKN1B, EZH2, NBN, DIS3, and MDM4) and 2 genes (FBXW7 and AURKA) were associated with decreased or increased risk of local recurrence, respectively (q-value ≤ 0.10). Weighted scores corresponding to the strength of association with local failure for each gene were summed to calculate a patient-level RPS. On multivariable Fine/Gray's competing risks regression, RPS [1.66 (1.44-1.91, P < .001)], metastasis-associated edema [1.60 (1.16-2.21), P = .004], baseline size [1.02 (1.01-1.03), P < .001] and receipt of WBRT without local therapy [4.04 (2.49-6.58), P < .001] were independent predictors of local failure. CONCLUSIONS We developed a genomic score to quantify local recurrence risk following brain-directed radiation. To the best of our knowledge, this represents the first study to systematically correlate DNA-based alterations with radiotherapeutic outcomes in BrM. If validated, Brain-RPS has potential to facilitate clinical trials aimed at genome-based personalization of radiation in BrM.
Collapse
Affiliation(s)
- Nayan Lamba
- Harvard Radiation Oncology Program, Harvard University, Boston, Massachusetts, USA
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | | | - Paul J Catalano
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, and Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Dewey Kim
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Hesham Elhalawani
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Daphne A Haas-Kogan
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women’s Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Nikhil Wagle
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Ayal A Aizer
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
31
|
Restifo D, McDermott JR, Cvetkovic D, Santos TD, Ogier C, Surumbayeva A, Handorf EA, Schimke C, Ma C, Cai KQ, Olszanski AJ, Kathad U, Bhatia K, Sharma P, Kulkarni A, Astsaturov I. Conditional Dependency of LP-184 on Prostaglandin Reductase 1 is Synthetic Lethal in Pancreatic Cancers with DNA Damage Repair Deficiencies. Mol Cancer Ther 2023; 22:1182-1190. [PMID: 37552607 PMCID: PMC10592171 DOI: 10.1158/1535-7163.mct-22-0818] [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: 12/27/2022] [Revised: 03/22/2023] [Accepted: 08/01/2023] [Indexed: 08/10/2023]
Abstract
The greater efficacy of DNA-damaging drugs for pancreatic adenocarcinoma (PDAC) relies on targeting cancer-specific vulnerabilities while sparing normal organs and tissues due to their inherent toxicities. We tested LP-184, a novel acylfulvene analog, for its activity in preclinical models of PDAC carrying mutations in the DNA damage repair (DDR) pathways. Cytotoxicity of LP-184 is solely dependent on prostaglandin reductase 1 (PTGR1), so that PTGR1 expression robustly correlates with LP-184 cytotoxicity in vitro and in vivo. Low-passage patient-derived PDAC xenografts with DDR deficiencies treated ex vivo are more sensitive to LP-184 compared with DDR-proficient tumors. Additional in vivo testing of PDAC xenografts for their sensitivity to LP-184 demonstrates marked tumor growth inhibition in models harboring pathogenic mutations in ATR, BRCA1, and BRCA2. Depletion of PTGR1, however, completely abrogates the antitumor effect of LP-184. Testing combinatorial strategies for LP-184 aimed at deregulation of nucleotide excision repair proteins ERCC3 and ERCC4 established synergy. Our results provide valuable biomarkers for clinical testing of LP-184 in a large subset of genetically defined characterized refractory carcinomas. High PTGR1 expression and deleterious DDR mutations are present in approximately one third of PDAC making these patients ideal candidates for clinical trials of LP-184.
Collapse
Affiliation(s)
- Diana Restifo
- The Marvin & Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | | | - Dusica Cvetkovic
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Troy Dos Santos
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Charline Ogier
- The Marvin & Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Aizhan Surumbayeva
- The Marvin & Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | | | | | - Charlie Ma
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Kathy Q. Cai
- Histopathology Facility, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Anthony J. Olszanski
- Department of Hematology and Oncology, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | | | | | | | | | - Igor Astsaturov
- The Marvin & Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
- Department of Hematology and Oncology, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| |
Collapse
|
32
|
Baird L, Taguchi K, Zhang A, Takahashi Y, Suzuki T, Kensler TW, Yamamoto M. A NRF2-induced secretory phenotype activates immune surveillance to remove irreparably damaged cells. Redox Biol 2023; 66:102845. [PMID: 37597423 PMCID: PMC10458321 DOI: 10.1016/j.redox.2023.102845] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 08/21/2023] Open
Abstract
While it is well established that the KEAP1-NRF2 pathway regulates the main inducible cellular response to oxidative stress, this cytoprotective function of NRF2 could become deleterious to the host if it confers survival onto irreparably damaged cells. In this regard, we have found that in diseased states, NRF2 promotes the transcriptional activation of a specific subset of the senescence-associated secretory phenotype (SASP) gene program, which we have named the NRF2-induced secretory phenotype (NISP). In two models of hepatic disease using Pten::Keap1 and Keap1::Atg7 double knockout mice, we found that the NISP functions in the liver to recruit CCR2 expressing monocytes, which function as immune system effector cells to directly remove the damaged cells. Through activation of this immune surveillance pathway, in non-transformed cells, NRF2 functions as a tumour suppressor to mitigate the long-term survival of damaged cells which otherwise would be detrimental for host survival. This pathway represents the final stage of the oxidative stress response, as it allows cells to be safely removed if the macromolecular damage caused by the original stressor is so extensive that it is beyond the repair capacity of the cell.
Collapse
Affiliation(s)
- Liam Baird
- Department of Biochemistry and Molecular Biology, Tohoku University, Tohoku Medical Megabank Organization, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan; Advanced Research Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, Sendai, 980-8575, Japan.
| | - Keiko Taguchi
- Department of Biochemistry and Molecular Biology, Tohoku University, Tohoku Medical Megabank Organization, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
| | - Anqi Zhang
- Department of Biochemistry and Molecular Biology, Tohoku University, Tohoku Medical Megabank Organization, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
| | - Yushi Takahashi
- Department of Biochemistry and Molecular Biology, Tohoku University, Tohoku Medical Megabank Organization, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
| | - Takafumi Suzuki
- Department of Biochemistry and Molecular Biology, Tohoku University, Tohoku Medical Megabank Organization, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
| | - Thomas W Kensler
- Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, United States
| | - Masayuki Yamamoto
- Department of Biochemistry and Molecular Biology, Tohoku University, Tohoku Medical Megabank Organization, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan; Advanced Research Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, Sendai, 980-8575, Japan.
| |
Collapse
|
33
|
Cui J, Guo Y, Yin T, Gou S, Xiong J, Liang X, Lu C, Peng T. USP8 promotes gemcitabine resistance of pancreatic cancer via deubiquitinating and stabilizing Nrf2. Biomed Pharmacother 2023; 166:115359. [PMID: 37639742 DOI: 10.1016/j.biopha.2023.115359] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 08/31/2023] Open
Abstract
Gemcitabine (Gem) is the first-line chemotherapy drug for pancreatic cancer, but the acquired chemoresistance also hinders its application. Therefore, research about Gem resistance plays a crucial role in enhancing the therapeutic effect of Gem. As a deubiquitinating enzyme, ubiquitin-specific protease 8 (USP8) was shown to play vital roles in the tumorigenesis processes of several cancers; however, the effect of USP8 on Gem resistance of pancreatic cancer still remains largely unknown. In the current study, we observed that the expression of USP8 was increased in pancreatic cancer patients, it is related to the recurrence of Gem chemotherapy, and USP8 expression could be induced by Gem application. Furthermore, USP8 was found to promote Gem resistance both in vivo and in vitro via regulating cell viability and apoptosis. Moreover, USP8 enhanced the activation of Nrf2 signaling which is dependent on its deubiquitinase ability. At last, we illustrated that USP8 interacted with Nrf2 directly and deubiquitinated K48-linked polyubiquitin chains from Nrf2, stabilizing the expression of Nrf2. In summary, the manuscript revealed the role of USP8 in Gem chemoresistance and suggested USP8 as a potential therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Jing Cui
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yao Guo
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tao Yin
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shanmiao Gou
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiongxin Xiong
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xueyi Liang
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chong Lu
- Department of Thyroid and Breast Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Tao Peng
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
34
|
Bitterman DS, Gensheimer MF, Jaffray D, Pryma DA, Jiang SB, Morin O, Ginart JB, Upadhaya T, Vallis KA, Buatti JM, Deasy J, Hsiao HT, Chung C, Fuller CD, Greenspan E, Cloyd-Warwick K, Courdy S, Mao A, Barnholtz-Sloan J, Topaloglu U, Hands I, Maurer I, Terry M, Curran WJ, Le QT, Nadaf S, Kibbe W. Cancer Informatics for Cancer Centers: Sharing Ideas on How to Build an Artificial Intelligence-Ready Informatics Ecosystem for Radiation Oncology. JCO Clin Cancer Inform 2023; 7:e2300136. [PMID: 38055914 PMCID: PMC10703125 DOI: 10.1200/cci.23.00136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/15/2023] [Accepted: 10/16/2023] [Indexed: 12/08/2023] Open
Abstract
In August 2022, the Cancer Informatics for Cancer Centers brought together cancer informatics leaders for its biannual symposium, Precision Medicine Applications in Radiation Oncology, co-chaired by Quynh-Thu Le, MD (Stanford University), and Walter J. Curran, MD (GenesisCare). Over the course of 3 days, presenters discussed a range of topics relevant to radiation oncology and the cancer informatics community more broadly, including biomarker development, decision support algorithms, novel imaging tools, theranostics, and artificial intelligence (AI) for the radiotherapy workflow. Since the symposium, there has been an impressive shift in the promise and potential for integration of AI in clinical care, accelerated in large part by major advances in generative AI. AI is now poised more than ever to revolutionize cancer care. Radiation oncology is a field that uses and generates a large amount of digital data and is therefore likely to be one of the first fields to be transformed by AI. As experts in the collection, management, and analysis of these data, the informatics community will take a leading role in ensuring that radiation oncology is prepared to take full advantage of these technological advances. In this report, we provide highlights from the symposium, which took place in Santa Barbara, California, from August 29 to 31, 2022. We discuss lessons learned from the symposium for data acquisition, management, representation, and sharing, and put these themes into context to prepare radiation oncology for the successful and safe integration of AI and informatics technologies.
Collapse
Affiliation(s)
- Danielle S. Bitterman
- Artificial Intelligence in Medicine (AIM) Program, Mass General Brigham, Harvard Medical School, Boston, MA
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Boston, MA
| | - Michael F. Gensheimer
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - David Jaffray
- Department of Radiation Physics, M.D. Anderson Cancer Center, Houston, TX
| | - Daniel A. Pryma
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Steve B. Jiang
- Medical Artificial Intelligence and Automation Laboratory and Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Olivier Morin
- Department of Radiation Oncology, MEDomics Laboratory, University of California San Francisco, San Francisco, CA
| | - Jorge Barrios Ginart
- Department of Radiation Oncology, MEDomics Laboratory, University of California San Francisco, San Francisco, CA
| | - Taman Upadhaya
- Department of Radiation Oncology, MEDomics Laboratory, University of California San Francisco, San Francisco, CA
| | - Katherine A. Vallis
- Artificial Intelligence in Medicine (AIM) Program, Mass General Brigham, Harvard Medical School, Boston, MA
| | - John M. Buatti
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Joseph Deasy
- Department of Radiation Oncology, University of Iowa Carver College of Medicine, Iowa City, IA
| | - H. Timothy Hsiao
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Caroline Chung
- Department of Scientific Affairs, American Society for Radiation Oncology, Arlington, VA
| | - Clifton D. Fuller
- Department of Scientific Affairs, American Society for Radiation Oncology, Arlington, VA
| | - Emily Greenspan
- Department of Radiation Oncology, M.D. Anderson Cancer Center, Houston, TX
| | - Kristy Cloyd-Warwick
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Rockville, MD
| | | | | | - Jill Barnholtz-Sloan
- Department of Radiation Oncology, M.D. Anderson Cancer Center, Houston, TX
- Center for Informatics, Digital Vertical, City of Hope National Comprehensive Cancer Center, Los Angeles, CA
| | - Umit Topaloglu
- Department of Radiation Oncology, M.D. Anderson Cancer Center, Houston, TX
| | - Isaac Hands
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
- Cancer Research Informatics Shared Resource Facility, University of Kentucky Markey Cancer Center, Lexington, NY
| | | | | | | | - Quynh-Thu Le
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Sorena Nadaf
- Department of Radiation Oncology, Emory University, Atlanta, GA
| | - Warren Kibbe
- Cancer Center Informatics Society, Los Angeles, CA
| |
Collapse
|
35
|
Wei YC, Zhu JY, Wu J, Yu S, Li W, Zhu MX, Liu TS, Cui YH, Li Q. Nestin overexpression reduces the sensitivity of gastric cancer cells to trastuzumab. J Gastrointest Oncol 2023; 14:1694-1706. [PMID: 37720426 PMCID: PMC10502550 DOI: 10.21037/jgo-22-1048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 07/21/2023] [Indexed: 09/19/2023] Open
Abstract
Background Trastuzumab (TRA) shows significant efficacy in patients with human epidermal growth factor receptor 2 (HER2)-positive gastric cancer (GC). While TRA can help treat HER2-positive breast cancer, TRA resistance is a key clinical challenge. Nestin reportedly regulates the cellular redox homeostasis in lung cancer. This study aimed at identifying the functions of Nestin on the TRA sensitivity of HER2-positive GC cells. Methods Real-time polymerase chain reaction (PCR) and Western blotting (WB) were performed to explore the association between the mRNA and protein expression profiles, respectively, of Nestin and the Keap1-Nrf2 pathway. The influence of Nestin overexpression on the in vitro sensitivity of GC cells to TRA was explored by Cell Counting Kit-8 (CCK-8) assay, colony formation assay, reactive oxygen species (ROS) detection, and flow cytometry. Results TRA treatment caused Nestin downregulation in two HER2-positive GC cell lines (MKN45 and NCI-N87). Nestin overexpression reduced the sensitivity of GC cells to TRA. The expression and activity of Nrf2 and relevant downstream antioxidant genes were increased by Nestin overexpression. Nestin overexpression also significantly suppressed TRA-induced apoptosis and ROS generation. In vivo tumor growth experiment with female BALB/c nude mice indicated that Nestin upregulation restored the tumor growth rate which was inhibited by TRA treatment. Conclusions Collectively, the inhibitory effect of Nestin on the TRA sensitivity of cells to TRA was confirmed in this study. These results imply that the antioxidant Nestin-Nrf2 axis may play a role in the mechanism underlying the resistance of GC cells to TRA.
Collapse
Affiliation(s)
- Yi-Chou Wei
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiang-Yi Zhu
- Department of Radiotherapy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Wu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shan Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Meng-Xuan Zhu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tian-Shu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yue-Hong Cui
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qian Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
36
|
Tanaka I, Koyama J, Itoigawa H, Hayai S, Morise M. Metabolic barriers in non-small cell lung cancer with LKB1 and/or KEAP1 mutations for immunotherapeutic strategies. Front Oncol 2023; 13:1249237. [PMID: 37675220 PMCID: PMC10477992 DOI: 10.3389/fonc.2023.1249237] [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: 06/28/2023] [Accepted: 08/08/2023] [Indexed: 09/08/2023] Open
Abstract
Currently, immune checkpoint inhibitors (ICIs) are widely considered the standard initial treatment for advanced non-small cell lung cancer (NSCLC) when there are no targetable driver oncogenic alternations. NSCLC tumors that have two alterations in tumor suppressor genes, such as liver kinase B1 (LKB1) and/or Kelch-like ECH-associated protein 1 (KEAP1), have been found to exhibit reduced responsiveness to these therapeutic strategies, as revealed by multiomics analyses identifying immunosuppressed phenotypes. Recent advancements in various biological approaches have gradually unveiled the molecular mechanisms underlying intrinsic reprogrammed metabolism in tumor cells, which contribute to the evasion of immune responses by the tumor. Notably, metabolic alterations in glycolysis and glutaminolysis have a significant impact on tumor aggressiveness and the remodeling of the tumor microenvironment. Since glucose and glutamine are essential for the proliferation and activation of effector T cells, heightened consumption of these nutrients by tumor cells results in immunosuppression and resistance to ICI therapies. This review provides a comprehensive summary of the clinical efficacies of current therapeutic strategies against NSCLC harboring LKB1 and/or KEAP1 mutations, along with the metabolic alterations in glycolysis and glutaminolysis observed in these cancer cells. Furthermore, ongoing trials targeting these metabolic alterations are discussed as potential approaches to overcome the extremely poor prognosis associated with this type of cancer.
Collapse
Affiliation(s)
- Ichidai Tanaka
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | |
Collapse
|
37
|
Ling R, Wang J, Fang Y, Yu Y, Su Y, Sun W, Li X, Tang X. HDAC-an important target for improving tumor radiotherapy resistance. Front Oncol 2023; 13:1193637. [PMID: 37503317 PMCID: PMC10368992 DOI: 10.3389/fonc.2023.1193637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/28/2023] [Indexed: 07/29/2023] Open
Abstract
Radiotherapy is an important means of tumor treatment, but radiotherapy resistance has been a difficult problem in the comprehensive treatment of clinical tumors. The mechanisms of radiotherapy resistance include the repair of sublethal damage and potentially lethal damage of tumor cells, cell repopulation, cell cycle redistribution, and reoxygenation. These processes are closely related to the regulation of epigenetic modifications. Histone deacetylases (HDACs), as important regulators of the epigenetic structure of cancer, are widely involved in the formation of tumor radiotherapy resistance by participating in DNA damage repair, cell cycle regulation, cell apoptosis, and other mechanisms. Although the important role of HDACs and their related inhibitors in tumor therapy has been reviewed, the relationship between HDACs and radiotherapy has not been systematically studied. This article systematically expounds for the first time the specific mechanism by which HDACs promote tumor radiotherapy resistance in vivo and in vitro and the clinical application prospects of HDAC inhibitors, aiming to provide a reference for HDAC-related drug development and guide the future research direction of HDAC inhibitors that improve tumor radiotherapy resistance.
Collapse
Affiliation(s)
- Rui Ling
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jingzhi Wang
- Department of Radiotherapy Oncology, Affiliated Yancheng First Hospital of Nanjing University Medical School, First People’s Hospital of Yancheng, Yancheng, China
| | - Yuan Fang
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yunpeng Yu
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yuting Su
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Wen Sun
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiaoqin Li
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiang Tang
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
38
|
Park JM, Kim S, Bae SU, Byun SJ, Seo I, Lee HW. Nuclear Factor Erythroid 2-Related Factor 2/Kelch-Like ECH-Associated Protein 1 as a Predictor of Prognosis and Radiotherapy Resistance in Patients With Locally Advanced Rectal Cancer: A Prospective Analysis. J Korean Med Sci 2023; 38:e200. [PMID: 37401495 DOI: 10.3346/jkms.2023.38.e200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 04/16/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND The nuclear factor erythroid 2-related factor 2/Kelch-like ECH-associated protein 1 (Nrf2/Keap1) signaling pathway is involved in the regulation of cellular responses to oxidative stress. Nrf2 acts as a cell protector from inflammation, cellular damage, and tumorigenesis, whereas Keap1 is a negative regulator of Nrf2. Dysregulation of the Nrf2/Keap1 pathway results in tumorigenesis and the active metabolism of tumor cells, leading to high resistance to radiotherapy. This study aimed to evaluate the predictive role of Nrf2 and Keap1 in the radiosensitivity and prognosis of locally advanced rectal cancer (LARC). METHODS In total, 90 patients with LARC underwent surgery after preoperative chemoradiotherapy (CRT). Endoscopic biopsies from the tumors were obtained before radiation, and the Nrf2 and Keap1 expressions were assessed by immunohistochemistry. The response to therapy was evaluated after surgery following CRT according to the pathologic tumor regression grade. The disease-free survival (DFS) and overall survival rates were also documented. The association between the Nrf2 and Keap1 immunoreactivity and the clinicopathological parameters was analyzed. RESULTS The overexpression of the nuclear Nrf2 before CRT showed a significant correlation with better DFS. The cytoplasmic Nrf2 expression was associated with more residual tumors after radiotherapy and a more unfavorable DFS, indicating lower radiosensitivity. CONCLUSION CRT is an important issue in LARC and is a major aspect of treatment. Thus, the Nrf2/Keap1 expression may be a potential predictor of preoperative therapeutic resistance. The Nrf2-Keap1 modulators that interact with each other may also be effectively applicable to CRT effect in LARC.
Collapse
Affiliation(s)
- Ji Min Park
- Department of Pathology, Keimyung University Dongsan Hospital, Daegu, Korea
| | - Shin Kim
- Department of Immunology, Keimyung University School of Medicine, Daegu, Korea
- Institute of Medical Science, Keimyung University, Daegu, Korea
- Institute for Cancer Research, Keimyung University, Daegu, Korea
| | - Sung Uk Bae
- Institute of Medical Science, Keimyung University, Daegu, Korea
- Institute for Cancer Research, Keimyung University, Daegu, Korea
- Department of Surgery, Keimyung University Dongsan Hospital, Daegu, Korea
| | - Sang Jun Byun
- Department Radiation Oncology, Keimyung University School of Medicine, Daegu, Korea
| | - Incheol Seo
- Department of Immunology, School of Medicine, Kyungpook National University, Daegu, Korea.
| | - Hye Won Lee
- Department of Pathology, Keimyung University Dongsan Hospital, Daegu, Korea
- Institute for Cancer Research, Keimyung University, Daegu, Korea.
| |
Collapse
|
39
|
Earland N, Chen K, Semenkovich NP, Chauhan PS, Zevallos JP, Chaudhuri AA. Emerging Roles of Circulating Tumor DNA for Increased Precision and Personalization in Radiation Oncology. Semin Radiat Oncol 2023; 33:262-278. [PMID: 37331781 DOI: 10.1016/j.semradonc.2023.03.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Recent breakthroughs in circulating tumor DNA (ctDNA) technologies present a compelling opportunity to combine this emerging liquid biopsy approach with the field of radiogenomics, the study of how tumor genomics correlate with radiotherapy response and radiotoxicity. Canonically, ctDNA levels reflect metastatic tumor burden, although newer ultrasensitive technologies can be used after curative-intent radiotherapy of localized disease to assess ctDNA for minimal residual disease (MRD) detection or for post-treatment surveillance. Furthermore, several studies have demonstrated the potential utility of ctDNA analysis across various cancer types managed with radiotherapy or chemoradiotherapy, including sarcoma and cancers of the head and neck, lung, colon, rectum, bladder, and prostate . Additionally, because peripheral blood mononuclear cells are routinely collected alongside ctDNA to filter out mutations associated with clonal hematopoiesis, these cells are also available for single nucleotide polymorphism analysis and could potentially be used to detect patients at high risk for radiotoxicity. Lastly, future ctDNA assays will be utilized to better assess locoregional MRD in order to more precisely guide adjuvant radiotherapy after surgery in cases of localized disease, and guide ablative radiotherapy in cases of oligometastatic disease.
Collapse
Affiliation(s)
- Noah Earland
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO; Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| | - Kevin Chen
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| | - Nicholas P Semenkovich
- Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Pradeep S Chauhan
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| | - Jose P Zevallos
- Department of Otolaryngology, University of Pittsburgh Medical School, Pittsburgh, PA
| | - Aadel A Chaudhuri
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO; Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO; Siteman Cancer Center, Barnes Jewish Hospital and Washington University School of Medicine, St. Louis, MO; Department of Genetics, Washington University School of Medicine, St. Louis, MO; Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, MO; Department of Computer Science and Engineering, Washington University in St. Louis, St. Louis, MO.
| |
Collapse
|
40
|
DeBlasi JM, Falzone A, Caldwell S, Prieto-Farigua N, Prigge JR, Schmidt EE, Chio IIC, Karreth FA, DeNicola GM. Distinct Nrf2 Signaling Thresholds Mediate Lung Tumor Initiation and Progression. Cancer Res 2023; 83:1953-1967. [PMID: 37062029 PMCID: PMC10267679 DOI: 10.1158/0008-5472.can-22-3848] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/03/2023] [Accepted: 04/12/2023] [Indexed: 04/17/2023]
Abstract
Mutations in the KEAP1-NRF2 (Kelch-like ECH-associated protein 1-nuclear factor-erythroid 2 p45-related factor 2) pathway occur in up to a third of non-small cell lung cancer (NSCLC) cases and often confer resistance to therapy and poor outcomes. Here, we developed murine alleles of the KEAP1 and NRF2 mutations found in human NSCLC and comprehensively interrogated their impact on tumor initiation and progression. Chronic NRF2 stabilization by Keap1 or Nrf2 mutation was not sufficient to induce tumorigenesis, even in the absence of tumor suppressors, p53 or LKB1. When combined with KrasG12D/+, constitutive NRF2 activation promoted lung tumor initiation and early progression of hyperplasia to low-grade tumors but impaired their progression to advanced-grade tumors, which was reversed by NRF2 deletion. Finally, NRF2 overexpression in KEAP1 mutant human NSCLC cell lines was detrimental to cell proliferation, viability, and anchorage-independent colony formation. Collectively, these results establish the context-dependence and activity threshold for NRF2 during the lung tumorigenic process. SIGNIFICANCE Stabilization of the transcription factor NRF2 promotes oncogene-driven tumor initiation but blocks tumor progression, indicating distinct, threshold-dependent effects of the KEAP1/NRF2 pathway in different stages of lung tumorigenesis.
Collapse
Affiliation(s)
- Janine M. DeBlasi
- Department of Metabolism & Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Cancer Biology PhD Program, University of South Florida, Tampa, Florida
| | - Aimee Falzone
- Department of Metabolism & Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Samantha Caldwell
- Department of Metabolism & Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Nicolas Prieto-Farigua
- Department of Metabolism & Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Justin R. Prigge
- Microbiology & Cell Biology Department, Montana State University, Bozeman, Montana
| | - Edward E. Schmidt
- Microbiology & Cell Biology Department, Montana State University, Bozeman, Montana
| | - Iok In Christine Chio
- Department of Genetics and Development, Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, New York
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Florian A. Karreth
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Gina M. DeNicola
- Department of Metabolism & Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
41
|
Wu WY, Jiao X, Song WX, Wu P, Xiao PQ, Huang XF, Wang K, Zhan SF. Network pharmacology and bioinformatics analysis identifies potential therapeutic targets of Naringenin against COVID-19/LUSC. Front Endocrinol (Lausanne) 2023; 14:1187882. [PMID: 37347115 PMCID: PMC10281056 DOI: 10.3389/fendo.2023.1187882] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) is a highly contagious respiratory disease that has posed a serious threat to people's daily lives and caused an unprecedented challenge to public health and people's health worldwide. Lung squamous cell carcinoma (LUSC) is a common type of lung malignancy with a highly aggressive nature and poor prognosis. Patients with LUSC could be at risk for COVID-19, We conducted this study to examine the potential for naringenin to develop into an ideal medicine and investigate the underlying action mechanisms of naringenin in COVID-19 and LUSC due to the anti-viral, anti-tumor, and anti-inflammatory activities of naringenin. Methods LUSC related genes were obtained from TCGA, PharmGKB, TTD,GeneCards and NCBI, and then the transcriptome data for COVID-19 was downloaded from GEO, DisGeNET, CTD, DrugBank, PubChem, TTD, NCBI Gene, OMIM. The drug targets of Naringenin were revealed through CTD, BATMAN, TCMIP, SymMap, Chemical Association Networks, SwissTargetPrediction, PharmMapper, ECTM, and DGIdb. The genes related to susceptibility to COVID-19 in LUSC patients were obtained through differential analysis. The interaction of COVID-19/LUSC related genes was evaluated and demonstrated using STRING to develop a a COX risk regression model to screen and evaluate the association of genes with clinical characteristics. To investigate the related functional and pathway analysis of the common targets of COVID-19/LUSC and Naringenin, KEGG and GO enrichment analysis were employed to perform the functional analysis of the target genes. Finally, The Hub Gene was screened and visualized using Cytoscape, and molecular docking between the drug and the target was performed using Autodock. Results We discovered numerous COVID-19/LUSC target genes and examined their prognostic value in LUSC patients utilizing a variety of bioinformatics and network pharmacology methods. Furthermore, a risk score model with strong predictive performance was developed based on these target genes to assess the prognosis of LUSC patients with COVID-19. We intersected the therapeutic target genes of naringenin with the LUSC, COVID-19-related targets, and identified 354 common targets, which could be used as potential target genes for naringenin to treat COVID-19/LUSC. The treatment of COVID-19/LUSC with naringenin may involve oxidative stress, anti-inflammatory, antiviral, antiviral, apoptosis, immunological, and multiple pathways containing PI3K-Akt, HIF-1, and VEGF, according to the results of the GO and KEGG enrichment analysis of these 354 common targets. By constructing a PPI network, we ascertained AKT1, TP53, SRC, MAPK1, MAPK3, and HSP90AA1 as possible hub targets of naringenin for the treatment of COVID-19/LUSC. Last but not least, molecular docking investigations showed that naringenin has strong binding activity in COVID-19/LUSC. Conclusion We revealed for the first time the pharmacological targets and potential molecular processes of naringenin for the treatment of COVID-19/LUSC. However, these results need to be confirmed by additional research and validation in real LUSC patients with COVID-19.
Collapse
Affiliation(s)
- Wen-yu Wu
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xin Jiao
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wen-xin Song
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peng Wu
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Pei-qi Xiao
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiu-fang Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kai Wang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shao-feng Zhan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
42
|
Lyu F, Shang SY, Gao XS, Ma MW, Xie M, Ren XY, Liu MZ, Chen JY, Li SS, Huang L. Uncovering the Secrets of Prostate Cancer's Radiotherapy Resistance: Advances in Mechanism Research. Biomedicines 2023; 11:1628. [PMID: 37371723 PMCID: PMC10296152 DOI: 10.3390/biomedicines11061628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/20/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Prostate cancer (PCa) is a critical global public health issue with its incidence on the rise. Radiation therapy holds a primary role in PCa treatment; however, radiation resistance has become increasingly challenging as we uncover more about PCa's pathogenesis. Our review aims to investigate the multifaceted mechanisms underlying radiation therapy resistance in PCa. Specifically, we will examine how various factors, such as cell cycle regulation, DNA damage repair, hypoxic conditions, oxidative stress, testosterone levels, epithelial-mesenchymal transition, and tumor stem cells, contribute to radiation therapy resistance. By exploring these mechanisms, we hope to offer new insights and directions towards overcoming the challenges of radiation therapy resistance in PCa. This can also provide a theoretical basis for the clinical application of novel ultra-high-dose-rate (FLASH) radiotherapy in the era of PCa.
Collapse
Affiliation(s)
- Feng Lyu
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China; (F.L.); (S.-Y.S.); (M.-W.M.); (M.X.); (X.-Y.R.); (M.-Z.L.); (J.-Y.C.); (S.-S.L.); (L.H.)
| | - Shi-Yu Shang
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China; (F.L.); (S.-Y.S.); (M.-W.M.); (M.X.); (X.-Y.R.); (M.-Z.L.); (J.-Y.C.); (S.-S.L.); (L.H.)
- First Clinical Medical School, Hebei North University, Zhangjiakou 075000, China
| | - Xian-Shu Gao
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China; (F.L.); (S.-Y.S.); (M.-W.M.); (M.X.); (X.-Y.R.); (M.-Z.L.); (J.-Y.C.); (S.-S.L.); (L.H.)
| | - Ming-Wei Ma
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China; (F.L.); (S.-Y.S.); (M.-W.M.); (M.X.); (X.-Y.R.); (M.-Z.L.); (J.-Y.C.); (S.-S.L.); (L.H.)
| | - Mu Xie
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China; (F.L.); (S.-Y.S.); (M.-W.M.); (M.X.); (X.-Y.R.); (M.-Z.L.); (J.-Y.C.); (S.-S.L.); (L.H.)
| | - Xue-Ying Ren
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China; (F.L.); (S.-Y.S.); (M.-W.M.); (M.X.); (X.-Y.R.); (M.-Z.L.); (J.-Y.C.); (S.-S.L.); (L.H.)
| | - Ming-Zhu Liu
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China; (F.L.); (S.-Y.S.); (M.-W.M.); (M.X.); (X.-Y.R.); (M.-Z.L.); (J.-Y.C.); (S.-S.L.); (L.H.)
| | - Jia-Yan Chen
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China; (F.L.); (S.-Y.S.); (M.-W.M.); (M.X.); (X.-Y.R.); (M.-Z.L.); (J.-Y.C.); (S.-S.L.); (L.H.)
| | - Shan-Shi Li
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China; (F.L.); (S.-Y.S.); (M.-W.M.); (M.X.); (X.-Y.R.); (M.-Z.L.); (J.-Y.C.); (S.-S.L.); (L.H.)
| | - Lei Huang
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China; (F.L.); (S.-Y.S.); (M.-W.M.); (M.X.); (X.-Y.R.); (M.-Z.L.); (J.-Y.C.); (S.-S.L.); (L.H.)
| |
Collapse
|
43
|
Guo SB, Du S, Cai KY, Cai HJ, Huang WJ, Tian XP. A scientometrics and visualization analysis of oxidative stress modulator Nrf2 in cancer profiles its characteristics and reveals its association with immune response. Heliyon 2023; 9:e17075. [PMID: 37342570 PMCID: PMC10277599 DOI: 10.1016/j.heliyon.2023.e17075] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/23/2023] Open
Abstract
Background Nrf2, an essential and fascinating transcription factor, enjoys a dual property in the occurrence and development of inflammation and cancer. For over two decades, numerous studies regarding Nrf2 in cancer have been reported, whereas there is still a lack of a scientometrics and visualization analysis of Nrf2 in cancer. Hence, a scientometric study regarding the oxidative stress modulator Nrf2 was implemented. Methods After the quality screening, we defined 7168 relevant studies from 2000 to 2021. CiteSpace, VOSviewer, R software, and GraphPad Prism were used for the following scientometric study and visualization analysis, including field profiles, research hotspots, and future predictions. Results The total number of publications and citations are 1058 and 54,690, respectively. After polynomial fitting curve analysis, two prediction functions of the annual publication number (y = 3.3909x2 - 13585x + 1 E+07) and citation number (185.45x2 - 743669x + 7 E+08) were generated. After scientometric analysis, we found that Biochemistry Molecular Biology correlates with Nrf2 in cancer highly, and Free Radical Biology and Medicine is a good choice for submitting Nrf2-related manuscripts. The current research hotspots of Nrf2 in cancer mainly focus on cancer therapy and its cellular and molecular mechanisms. "antioxidant response element (87.5)", "gene expression (43.98)", "antioxidant responsive element (21.14)", "chemoprevention (20.05)", "carcinogenesis (19.2)", "cancer chemoprevention (18.45)", "free radical (17.15)", "response element (14.17)", and "chemopreventive agent (14.04)" are important for cancer therapy study. In addition, "glutathione-S-transferase (47)", "keap1 (15.39)", and "heme oxygenase 1 gene (24.35)" are important for inflammation and cell fate study. More interestingly, by performing an "InfoMap" algorithm, the thematic map showed that the "immune response" is essential to oxidative stress modulator Nrf2 but not well developed, indicating it deserves further exploration. Conclusion This study revealed field profiles, research hotspots, and future directions of oxidative stress modulator Nrf2 in inflammation and cancer research, and our findings will offer a vigorous roadmap for further studies in this field.
Collapse
Affiliation(s)
- Song-Bin Guo
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Sheng Du
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, PR China
| | - Ke-Yu Cai
- Department of Colorectal and Anal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, PR China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, PR China
| | - Han-Jia Cai
- The Second Clinical Medical College, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Wei-Juan Huang
- Department of Pharmacology, College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Xiao-Peng Tian
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| |
Collapse
|
44
|
Allignet B, De Ruysscher D, Martel-Lafay I, Waissi W. Stereotactic body radiation therapy in unresectable stage III non-small cell lung cancer: A systematic review. Cancer Treat Rev 2023; 118:102573. [PMID: 37210766 DOI: 10.1016/j.ctrv.2023.102573] [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: 01/30/2023] [Revised: 03/29/2023] [Accepted: 05/11/2023] [Indexed: 05/23/2023]
Abstract
In unresectable stage III non-small cell lung cancer (NSCLC), the standard of care for most fit patients is concurrent chemotherapy with normofractionated radiotherapy (NFRT), followed by durvalumab consolidation. Nevertheless, almost half of patients will present locoregional or metastatic intrathoracic relapse. Improving locoregional control thus remains an important objective. For this purpose, stereotactic body radiotherapy (SBRT) may be a relevant treatment modality. We performed a systematic review of the literature that evaluate the efficacy and safety of SBRT in this situation, either instead of or in addition to NFRT. Among 1788 unique reports, 18 met the inclusion criteria. They included 447 patients and were mainly prospective (n = 10, including 5 phase 2 trials). In none, maintenance durvalumab was administered. Most reported SBRT boost after NFRT (n = 8), or definitive tumor and nodal SBRT (n = 7). Median OS varied from 10 to 52 months, due to the heterogeneity of the included populations and according to treatment regimen. The rate of severe side effects was low, with <5 % grade 5 toxicity, and mainly observed when mediastinal SBRT was performed without dose constraints to the proximal bronchovascular tree. It was suggested that a biologically effective dose higher than 112.3 Gy may increase locoregional control. SBRT for selected stage III NSCLC bears potential to improve loco-regional tumor control, but at present, this should only be done in prospective clinical trials.
Collapse
Affiliation(s)
- Benoît Allignet
- Department of Radiation Oncology, Centre Léon Bérard, 28 rue Laennec, 69673 Lyon, France; Univ Lyon, INSA-Lyon, Université Claude Bernard Lyon 1, UJM-Saint Etienne, CNRS, Inserm, CREATIS UMR 5220, U1294 Lyon, France.
| | - Dirk De Ruysscher
- Department of Radiation Oncology (Maastro), Maastricht University Medical Center, GROW School for Oncology and Developmental Biology, The Netherlands; Department of Radiotherapy, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Isabelle Martel-Lafay
- Department of Radiation Oncology, Centre Léon Bérard, 28 rue Laennec, 69673 Lyon, France
| | - Waisse Waissi
- Department of Radiation Oncology, Centre Léon Bérard, 28 rue Laennec, 69673 Lyon, France
| |
Collapse
|
45
|
Hanley R, Pagliari F, Garcia-Calderón D, Fernandes Guerreiro J, Genard G, Jansen J, Nisticò C, Marafioti MG, Tirinato L, Seco J. Radio-resistance of hypoxic tumors: exploring the effects of oxygen and x-ray radiation on non-small lung cancer cell lines. Radiat Oncol 2023; 18:81. [PMID: 37173741 PMCID: PMC10182694 DOI: 10.1186/s13014-023-02275-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 04/29/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Solid tumors are often riddled with hypoxic areas, which develops as a result of high proliferation. Cancer cells willingly adapt and thrive in hypoxia by activating complex changes which contributes to survival and enhanced resistance to treatments, such as photon radiation. Photon radiation primarily relies on oxygen for the production of reactive oxygen species to induce DNA damage. The present in-vitro study aimed at investigating the biochemical responses of hypoxic non-small cell lung cancer (NSCLC) cells, particularly the effects on the DNA damage repair systems contributing to more radioresistant phenotypes and their pro- and anti-oxidant potential, within the first 24 h post-IR. METHODS NSCLC cell lines (H460, A549, Calu-1) were irradiated using varying X-ray doses under normoxia (21% O2) and hypoxia (0.1% O2). The overall cell survival was assessed by clonogenic assays. The extent of irradiation (IR)-induced DNA damage was evaluated by analyzing γ-H2AX foci induction and the altered expression of repair genes involved in non-homologous end joining and homologous recombination pathways. Moreover, cell-altered responses were investigated, including the nuclear and cytosolic hydrogen peroxide (H2O2) production, as well as the associated anti-oxidant potential, in particular some components related to the glutathione system. RESULTS Analysis of clonogenic survival revealed an enhanced radioresistance of the hypoxic NSCLC cells associated with reduced DNA damage and a downregulation of DNA repair genes. Moreover, nuclear H2O2 levels were IR-induced in a dose-dependent manner only under normoxia, and directly correlated with the DNA double-strand breaks. However, the observed nuclear H2O2 reduction in hypoxia appeared to be unaffected by IR, thus highlighting a possible reason for the enhanced radioresistance of the hypoxic NSCLC cells. The cellular antioxidant capacity was upregulated by IR in both oxygen conditions most likely helping to counteract the radiation effect on the cytosolic H2O2. CONCLUSIONS In conclusion, our data provide insight into the adaptive behavior of radiation-resistant hypoxic NSCLC cells, in particular their DNA repair and oxidative stress responses, which could contribute to lower DNA damage and higher cell survival rates following X-ray exposure. These findings may therefore help to identify potential targets for improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Rachel Hanley
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld, Heidelberg, Germany
- Department of Physics and Astronomy, Heidelberg University, Im Neuenheimer Feld, Heidelberg, Germany
| | - Francesca Pagliari
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld, Heidelberg, Germany.
| | - Daniel Garcia-Calderón
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld, Heidelberg, Germany
- Department of Physics and Astronomy, Heidelberg University, Im Neuenheimer Feld, Heidelberg, Germany
| | - Joana Fernandes Guerreiro
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld, Heidelberg, Germany
- Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisbon, Portugal
| | - Géraldine Genard
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld, Heidelberg, Germany
| | - Jeannette Jansen
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld, Heidelberg, Germany
| | - Clelia Nisticò
- Experimental and Clinical Medicine Department, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Maria Grazia Marafioti
- Experimental and Clinical Medicine Department, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Luca Tirinato
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld, Heidelberg, Germany
- Experimental and Clinical Medicine Department, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Joao Seco
- Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld, Heidelberg, Germany.
- Department of Physics and Astronomy, Heidelberg University, Im Neuenheimer Feld, Heidelberg, Germany.
| |
Collapse
|
46
|
Pan L, Wu Q, Wang Y, Ma S, Zhang S. Characterization and mechanisms of radioresistant lung squamous cell carcinoma cell lines. Thorac Cancer 2023; 14:1239-1250. [PMID: 37028947 PMCID: PMC10175036 DOI: 10.1111/1759-7714.14865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 04/09/2023] Open
Abstract
BACKGROUND Radiotherapy is an important clinical treatment for patients with lung squamous cell carcinoma (LUSC), and resistance to radiotherapy is an important cause of recurrence and metastasis in LUSC. The aim of this study was to establish and explore the biological characteristics of radioresistant LUSC cells. MATERIALS AND METHODS The LUSC cell lines NCI-H2170 and NCI-H520 were irradiated (4 Gy × 15Fraction). Radiosensitivity, cell apoptosis, cell cycle, and DNA damage repair were measured by clonogenic survival assay, flow cytometry, immunofluorescence for γ-H2AX foci, and Comet assay, respectively. Activation of p-ATM(Ser1981), p-CHK2(Th68), p-DNA-PKcs (Ser2056), and Ku70/Ku80 was measured by western blot. Proteomics was used to explore the differential genes and enriched signaling pathways between radioresistant cell lines and parental lines. In vivo nude mouse xenograft experiments further verified the feasibility of the radioresistant LUSC cell lines. RESULTS After fractionated irradiation (total dose of 60 Gy), radioresistant cells had decreased radiosensitivity, increased G0/G1 phase arrest, enhanced DNA damage repair ability, and through the ATM/CHK2 and DNA-PKcs/Ku70 pathways regulated double strands break. The upregulated differential genes in radioresistant cell lines were mainly enriched in biological pathways such as cell migration and extracellular matrix (ECM)-receptor interaction. In vivo verification of decreased radiosensitivity of radioresistant cells CONCLUSIONS: Radioresistant LUSC cell lines were established by fractional radiotherapy, which regulates IR-induced DNA damage repair through ATM/CHK2 and DNA-PKcs/Ku70. Tandem Mass Tags (TMT) quantitative proteomics found that the biological process pathway of cell migration and ECM-receptor interaction are upregulated in LUSC radioresistant cells.
Collapse
Affiliation(s)
- Lifang Pan
- Department of the Fourth Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiong Wu
- Department of Integrated Chinese and Western Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Yuqing Wang
- Department of Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shenglin Ma
- Department of the Fourth Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shirong Zhang
- Department of Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
47
|
Workman S, Jabbour SK, Deek MP. A narrative review of genetic biomarkers in non-small cell lung cancer: an update and future perspectives. AME MEDICAL JOURNAL 2023; 8:6. [PMID: 37025121 PMCID: PMC10072845 DOI: 10.21037/amj-2022-01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background and Objective Lung cancer has long been the leading cause of cancer deaths in the United States. Lung cancer has a poor prognosis, and our understanding of who will maximally benefit from different therapies is incomplete. This article discusses genetic biomarkers that may help in this regard. Methods From origin until February 25, 2022, PubMed database was searched for terms "non-small cell lung cancer", "genomics" and "biomarker", with special attention paid to literature published within the past 10 years. Search was language restricted to English. Additional literature was identified through hand searches of the references of retrieved literature. Key Content and Findings The most robustly described biomarkers for non-small cell lung cancer (NSCLC) are assessment of specific gene mutations. These are currently used in clinical practice for both prediction and prognostication. Abnormal mutation status of STK11/LKB1 and KEAP1-NFE2L2 are associated with poor response to radiotherapy (RT), and STK11/LKB1 is further associated with resistance to PD-L1 immunotherapy. Abnormal TP53 is associated with decreased benefit from cisplatin in squamous cell carcinoma (SCC). In terms of prognostication, RB1 mutations are associated with decreased overall survival (OS) in NSCLC and KEAP1-NFE2L2 mutations are associated with increased local recurrence (LR).Additional work has focused on gene expression levels, as well as analysis of genetic factors and signaling molecules affecting the tumor microenvironment (TME). High levels of Rad51c and NFE2L2 are associated with resistance to chemotherapy, and high Rad51c levels are further associated with resistance to RT. High nuclear expression of β-catenin has additionally been associated with poor RT response. Further, there is increasing evidence that some long non-coding RNAs (lncRNAs) may play a crucial role in regulation of tumor radiosensitivity. Much of this work has had promising early results but will require further validation before routine clinical use. Finally, there is evidence that quantification of some signaling molecules and microRNAs (miRNAs) may have clinical utility in predicting adverse outcomes in RT. Conclusions An improved understanding of tumor genetics in NSCLC has led to the development of targeted therapies and improved prognostication. As more work is done in this field, more and more genetic biomarkers will become candidates for clinical use. Much work will be required to validate these findings in the clinical setting.
Collapse
Affiliation(s)
- Samuel Workman
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Matthew P Deek
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| |
Collapse
|
48
|
Guan L, Nambiar DK, Cao H, Viswanathan V, Kwok S, Hui AB, Hou Y, Hildebrand R, von Eyben R, Holmes BJ, Zhao J, Kong CS, Wamsley N, Zhang W, Major MB, Seol SW, Sunwoo JB, Hayes DN, Diehn M, Le QT. NFE2L2 Mutations Enhance Radioresistance in Head and Neck Cancer by Modulating Intratumoral Myeloid Cells. Cancer Res 2023; 83:861-874. [PMID: 36652552 PMCID: PMC10023320 DOI: 10.1158/0008-5472.can-22-1903] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 11/18/2022] [Accepted: 01/13/2023] [Indexed: 01/19/2023]
Abstract
Radiotherapy (RT) is one of the primary treatments of head and neck squamous cell carcinoma (HNSCC), which has a high-risk of locoregional failure (LRF). Presently, there is no reliable predictive biomarker of radioresistance in HNSCC. Here, we found that mutations in NFE2L2, which encodes Nrf2, are associated with a significantly higher rate of LRF in patients with oral cavity cancer treated with surgery and adjuvant (chemo)radiotherapy but not in those treated with surgery alone. Somatic mutation of NFE2L2 led to Nrf2 activation and radioresistance in HNSCC cells. Tumors harboring mutant Nrf2E79Q were substantially more radioresistant than tumors with wild-type Nrf2 in immunocompetent mice, whereas the difference was diminished in immunocompromised mice. Nrf2E79Q enhanced radioresistance through increased recruitment of intratumoral polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC) and reduction of M1-polarized macrophages. Treatment with the glutaminase inhibitor CB-839 overcame the radioresistance induced by Nrf2E79Q or Nrf2E79K. RT increased expression of PMN-MDSC-attracting chemokines, including CXCL1, CXLC3, and CSF3, in Nrf2E79Q-expressing tumors via the TLR4, which could be reversed by CB-839. This study provides insights into the impact of NFE2L2 mutations on radioresistance and suggests that CB-839 can increase radiosensitivity by switching intratumoral myeloid cells to an antitumor phenotype, supporting clinical testing of CB-839 with RT in HNSCC with NFE2L2 mutations. SIGNIFICANCE NFE2L2 mutations are predictive biomarkers of radioresistance in head and neck cancer and confer sensitivity to glutaminase inhibitors to overcome radioresistance.
Collapse
Affiliation(s)
- Li Guan
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Dhanya K. Nambiar
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Hongbin Cao
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Vignesh Viswanathan
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Shirley Kwok
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Angela B. Hui
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Yuan Hou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Rachel Hildebrand
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Rie von Eyben
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Brittany J. Holmes
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Junfei Zhao
- Department of Pathology and Cell Biology, Columbia University, New York, USA
| | - Christina S. Kong
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Nathan Wamsley
- Washington University in St. Louis, Department of Cell Biology and Physiology, St. Louis, MO, USA
| | - Weiruo Zhang
- Department of Biomedical Data Science, School of Medicine, Stanford University, Stanford, CA, USA
| | - Michael B. Major
- Washington University in St. Louis, Department of Cell Biology and Physiology, St. Louis, MO, USA
| | - Seung W. Seol
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John B. Sunwoo
- OHNS/Head &Neck Surgery Divisions, Stanford University School of Medicine, Stanford, California, USA
| | - D. Neil Hayes
- Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Maximilian Diehn
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Quynh-Thu Le
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
49
|
Clinical utility of liquid biopsy and integrative genomic profiling in early-stage and oligometastatic cancer patients treated with radiotherapy. Br J Cancer 2023; 128:857-876. [PMID: 36550207 PMCID: PMC9977775 DOI: 10.1038/s41416-022-02102-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Liquid biopsy and Integrative Genomic Profiling (IGP) are yet to be implemented into routine Radiation Oncology. Here we assess the utility of germline, tumour and circulating cell-free DNA-based genomic analyses for the clinical management of early-stage and oligometastatic cancer patients treated by precision radiotherapy. METHODS We performed germline, tissue- and liquid biopsy NGS panels on 50 early-stage/oligometastatic cancer patients undergoing radiotherapy. We also monitored ctDNA variants in serial liquid biopsies collected during radiotherapy and follow-up and evaluated the clinical utility of such comprehensive approach. RESULTS The integration of different genomic studies revealed that only 1/3 of the liquid biopsy variants are of tumour origin. Altogether, 55 tumour variants (affecting 3/4 of the patients) were considered potentially actionable (for treatment and prognosis), whereas potential follow-up biomarkers were identified in all cases. Germline cancer-predisposing variants were present in three patients, which would have not been eligible for hereditary cancer testing according to clinical guidelines. The presence of detectable ctDNA variants before radiotherapy was associated with progression-free survival both in oligometastatic patients and in those with early-stage. CONCLUSIONS IGP provides both valuable and actionable information for personalised decision-making in Radiation Oncology.
Collapse
|
50
|
Ji L, Moghal N, Zou X, Fang Y, Hu S, Wang Y, Tsao MS. The NRF2 antagonist ML385 inhibits PI3K-mTOR signaling and growth of lung squamous cell carcinoma cells. Cancer Med 2023; 12:5688-5702. [PMID: 36305267 PMCID: PMC10028163 DOI: 10.1002/cam4.5311] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/11/2022] [Accepted: 09/20/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Lung squamous cell carcinoma (LUSC) currently has limited therapeutic options because of the relatively few validated targets and the lack of clinical drugs for some of these targets. Although NRF2/NFE2L2 pathway activation commonly occurs in LUSC, NRF2 has predominantly been studied in other cancer models. Here, we investigated the function of NRF2 in LUSC, including in organoid models, and we explored the activity of a small molecule NRF2 inhibitor ML385, which has not previously been investigated in LUSC. METHODS We first explored the role of NRF2 signaling in LUSC cancer cell line and organoid proliferation through NRF2 knockdown or ML385 treatment, both in vivo and in vitro. Next, we performed Western blot and immunofluorescence assays to determine the effect of NRF2 inhibition on PI3K-mTOR signaling. Finally, we used cell viability and clonogenic assays to explore whether ML385 could sensitize LUSC cancer cells to PI3K inhibitors. RESULTS We find that downregulation of NRF2 signaling inhibited proliferation of LUSC cancer cell lines and organoids, both in vivo and in vitro. We also demonstrate that inhibition of NRF2 reduces PI3K-mTOR signaling, with two potential mechanisms being involved. Although NRF2 promotes AKT phosphorylation, it also acts downstream of AKT to increase RagD protein expression and recruitment of mTOR to lysosomes after amino acid stimulation. We also find that ML385 potentiates LUSC growth inhibition by a pan-PI3K inhibitor, which correlates with stronger inhibition of PI3K-mTOR signaling. CONCLUSIONS Our data provide additional support for NRF2 promoting LUSC growth through PI3K-mTOR activation and support development of NRF2 inhibitors for the treatment of LUSC.
Collapse
Affiliation(s)
- Lili Ji
- Department of Pathology, Key Laboratory of Microenvironment and Translational Cancer Research, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Nadeem Moghal
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Xinru Zou
- Department of Pathology, Key Laboratory of Microenvironment and Translational Cancer Research, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Yixuan Fang
- Department of Pathology, Key Laboratory of Microenvironment and Translational Cancer Research, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Shuning Hu
- Department of Pathology, Key Laboratory of Microenvironment and Translational Cancer Research, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Yuhui Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Ming Sound Tsao
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|